1
|
Singh R, Gaur SK, Nagar R, Kaul R. Insights into the different mechanisms of Autophagy and Apoptosis mediated by Morbilliviruses. Virology 2025; 603:110371. [PMID: 39742556 DOI: 10.1016/j.virol.2024.110371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/10/2024] [Accepted: 12/20/2024] [Indexed: 01/03/2025]
Abstract
Viruses are obligate intracellular parasites that have co-evolved with the host. During the course of evolution, viruses have acquired abilities to abrogate the host's immune responses by modulating the host proteins which play a pivotal role in various biological processes. One such process is the programmed cell death in virus-infected cells, which can occur via autophagy or apoptosis. Morbilliviruses are known to modulate both autophagy and apoptosis. Upon infecting a cell, the morbilliviruses can utilize autophagosomes as their nest and delay the host defense apoptotic response, and/or can promote apoptosis to escalate the virus dissemination. Moreover, there is an active interplay between these two pathways which eventually decides the fate of a virus-infected cell. Recent advances in our understanding of these processes provide a potential rationale to further explore morbilliviruses for therapeutic purposes.
Collapse
Affiliation(s)
- Rashmi Singh
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Sharad Kumar Gaur
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Rakhi Nagar
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Rajeev Kaul
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India.
| |
Collapse
|
2
|
Gaur SK, Jain J, Chaudhary Y, Kaul R. Insights into the mechanism of Morbillivirus induced immune suppression. Virology 2024; 600:110212. [PMID: 39232265 DOI: 10.1016/j.virol.2024.110212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/26/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
Viruses enter the host cell, and various strategies are employed to evade the host immune system. These include overcoming the various components of the immune system, including modulation of the physical and chemical barriers, non-specific innate response and specific adaptive immune response. Morbilliviruses impose immune modulation by utilizing various approaches including hindering antigen presentation to T-Helper (TH) cells, hematopoiesis and suppression of effector molecule activities. These viruses can also impede the early stages of T cell activation. Despite the availability of effective vaccines, morbilliviruses are still a significant threat to mankind. After infection, they also induce a state of immune suppression in the host. The molecular mechanisms employed by morbilliviruses to induce the state of immune suppression in the infected host are still being investigated. This review is an attempt to summarize insights into some of the strategies adopted by morbilliviruses to mediate immune modulation in the host.
Collapse
Affiliation(s)
- Sharad Kumar Gaur
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Juhi Jain
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Yash Chaudhary
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Rajeev Kaul
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India.
| |
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
|
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
|
5
|
Li J, Zhong C, Liao Z, Mao L, Li W, Sun M, Liu M, Ji X, Liu C, Xue T, Yang L, Zhang W. Bta-miR-98 Suppresses Replication of Caprine Parainfluenza Virus Type 3 Through Inhibiting Apoptosis by Targeting Caspase-3. Front Immunol 2020; 11:1575. [PMID: 32983081 PMCID: PMC7484655 DOI: 10.3389/fimmu.2020.01575] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 06/15/2020] [Indexed: 12/26/2022] Open
Abstract
Caprine parainfluenza virus type 3 (CPIV3) is an emerging respiratory pathogen that affects the sheep and goat industry in China and possibly other countries around the world. Accumulating evidence suggests that microRNAs play important roles in regulating virus-host interactions and can suppress or facilitate viral replication. In this study, we showed that CPIV3 infection induced apoptosis in Madin-Darby bovine kidney (MDBK) cells, as determined by morphological changes and flow cytometry. Caspase activity and the expression of pro-apoptotic genes further indicated that CPIV3 induced apoptosis by activating both the intrinsic and extrinsic pathways. We also demonstrated the involvement of bta-microRNA-98 (bta-miR-98) in regulating CPIV3-induced apoptosis. Bta-miR-98 was downregulated in MDBK cells infected with CPIV3. Overexpression of bta-miR-98 significantly decreased the activities of caspase-3, -8, and -9. Conversely, inhibition of bta-miR-98 had completely opposite effects. Furthermore, our data showed that bta-miR-98 markedly affected CPIV3 replication by regulating apoptosis. Importantly, we found that bta-miR-98 modulated CPIV3-induced apoptosis by targeting caspase-3, an effector of apoptosis. Collectively, our results may suggest that CPIV3 infection induced apoptosis and downregulated the levels of bta-miR-98, and this miRNA regulated viral replication through effected apoptosis. This study contributes to our understanding of the molecular mechanisms underlying CPIV3 pathogenesis.
Collapse
Affiliation(s)
- Jizong Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China.,School of Pharmacy, Linyi University, Linyi, China.,Institute of Life Sciences, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Chunyan Zhong
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China.,College of Animal Science, Guizhou University, Guiyang, China
| | - Zheng Liao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China.,College of Animal Science, Guizhou University, Guiyang, China
| | - Li Mao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China.,Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Wenliang Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China.,Institute of Life Sciences, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Min Sun
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China
| | - Maojun Liu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China.,Institute of Life Sciences, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Xinqin Ji
- College of Animal Science, Guizhou University, Guiyang, China
| | - Chuanmin Liu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China
| | - Tao Xue
- School of Pharmacy, Linyi University, Linyi, China
| | - Leilei Yang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China
| | - Wenwen Zhang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China
| |
Collapse
|
6
|
Çomakli S, Özdemir S, Değirmençay Ş. Canine distemper virus induces downregulation of GABA A,GABA B, and GAT1 expression in brain tissue of dogs. Arch Virol 2020; 165:1321-1331. [PMID: 32253618 DOI: 10.1007/s00705-020-04617-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/11/2020] [Indexed: 02/08/2023]
Abstract
The aim of the study was to determine the expression profiles of GABAA, GABAB, and GAT1 using RT-PCR and the immunoreactivity of GAT1 via immunohistochemical and immunofluorescence assays in CDV-infected brain tissue of dogs. For this purpose, dogs with CDV and dogs without CDV were selected. The mRNA transcript levels of GABAA, GABAB, and GAT1 were significantly downregulated in brain tissue in the CDV-infected group as compared with that in non-CDV-infected brain tissue in the control group (p < 0.01, p < 0.001). In addition, the immunoreactivity of GAT1 in CDV-infected brain tissue was significantly lower than in the uninfected group (p < 0.05). We conclude that one of the main causes of myoclonus in CDV infections may be the blockage of postsynaptic inhibition in neurons or a lack of metabolism of GABA. In addition, a GABA neurotransmission imbalance could play a role in demyelination in CDV infections.
Collapse
Affiliation(s)
- Selim Çomakli
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Selçuk Özdemir
- Department of Genetic, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Şükrü Değirmençay
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| |
Collapse
|
7
|
Zheng X, Zhu Y, Zhao Z, Yan L, Xu T, Wang X, He H, Xia X, Zheng W, Xue X. RNA sequencing analyses of gene expressions in a canine macrophages cell line DH82 infected with canine distemper virus. INFECTION GENETICS AND EVOLUTION 2020; 80:104206. [PMID: 31982604 DOI: 10.1016/j.meegid.2020.104206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/18/2020] [Accepted: 01/22/2020] [Indexed: 02/06/2023]
Abstract
Virulent morbillivirus infections, including Meals Virus (MeV) and Canine Distemper Virus (CDV), caused severe immune suppression and leukopenia, while attenuated vaccine strains developed protective host immune responses. However, the detailed molecular foundations of host antiviral responses were poorly characterized. In order to better understand the interactions between attenuated vaccine and host antiviral responses, the global gene expression changes in CDV-11-infected DH82 cells, a macrophage-derived cell line from canine, were investigated by transcriptomic analysis, and portions of results were confirmed with quantitative RT-PCR. The results exhibited that 372 genes significantly up-regulated (p < .01) and 119 genes were significantly down-regulated (p < .01) in CDV-infected macrophages DH82 at 48 h p.i.. The enriched functions of the significantly up-regulated (p < .01) genes were closely associated with interferon stimulated genes (ISGs), chemokine genes and pro-inflammatory factor genes. Gene ontology and pathway analysis of differentially expressed genes (DEGs) revealed that the most significantly involved pathways in CDV-infected DH82 cells were NF-κB and TNF signaling pathway, cytokine-cytokine receptor interaction, and pathogen associated molecular patterns (PAMPs), such as Toll-like, RIG-I-like and NOD-like receptor signalings. Thus, the findings indicated that pattern recognition receptors (PRRs) possibly mediated host innate and protective antiviral immune responses in CDV-11 infected DH82 cells.
Collapse
Affiliation(s)
- Xuexing Zheng
- Department of Virology, School of Public Health, Shandong University, Jinan 250012, China.
| | - Yelei Zhu
- Department of Virology, School of Public Health, Shandong University, Jinan 250012, China; Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China
| | - Zhongxin Zhao
- Department of Virology, School of Public Health, Shandong University, Jinan 250012, China
| | - Lina Yan
- Department of Virology, School of Public Health, Shandong University, Jinan 250012, China
| | - Tong Xu
- Department of Virology, School of Public Health, Shandong University, Jinan 250012, China
| | - Xianwei Wang
- College of Life Sciences, Shandong University, Qingdao 266237, China
| | - Hongbin He
- College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Xianzhu Xia
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun 130122, China
| | - Wenwen Zheng
- Department of Virology, School of Public Health, Shandong University, Jinan 250012, China
| | - Xianghong Xue
- Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun 130122, China.
| |
Collapse
|
8
|
Xue X, Zhu Y, Yan L, Wong G, Sun P, Zheng X, Xia X. Antiviral efficacy of favipiravir against canine distemper virus infection in vitro. BMC Vet Res 2019; 15:316. [PMID: 31477101 PMCID: PMC6720089 DOI: 10.1186/s12917-019-2057-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/21/2019] [Indexed: 12/18/2022] Open
Abstract
Background Canine distemper (CD) is an acute infectious disease with high morbidity rates caused by a highly contagious pathogen (Canine Morbillivirus, also known as canine distemper virus, CDV). CDV can infect a broad range of carnivores resulting in complex clinical signs. Currently, there is no effective method to treat for CDV infections. Favipiravir (T-705), a pyrazine derivative, was shown to be an effective antiviral drug against RNA viruses, acting on RNA-dependent RNA polymerase (RdRp). However, whether the T-705 has antiviral effects following CDV infection is unclear. Here, we investigated the antiviral effect of T-705 against CDV-3 and CDV-11 strains in Vero and DH82 cell lines. Results Our data demonstrated that T-705 significantly inhibited the replication of CDV-3 and CDV-11 in both Vero and DH82 cells at different concentrations, ranging from 2.441 μg/ml to 1250 μg/ml. Additionally, T-705 exhibited efficacious antiviral effects when administered at different time points after virus infection. Cytotoxicity tests showed a slight decline in viability in Vero cells after T-705 treatment, and no apparent cytotoxicity was detected in T-705 treated DH82 cells. Comparison of anti-CDV polyclonal serum only inhibition of CDV in supernatant, T-705 directly inhibited viral replication in cells, and indirectly reduced the amount of virions in supernatant. The combination application of T-705 and anti-CDV polyclonal serum exhibited a rapid and robust inhibition against virions in supernatant and virus replication in cells. Conclusions Our data strongly indicated that T-705 effectively inhibited viral replication following CDV infection in vitro, and could be a potential candidate for treatment for CD.
Collapse
Affiliation(s)
- Xianghong Xue
- Department of Virology, School of Public Health, Shandong University, Jinan, 250012, China.,Division of Infectious Diseases of Special Animal, Institute of Special Animal and Plant Sciences, The Chinese Academy of Agricultural Sciences, Changchun, 130112, China
| | - Yelei Zhu
- Department of Virology, School of Public Health, Shandong University, Jinan, 250012, China.,Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Lina Yan
- Department of Virology, School of Public Health, Shandong University, Jinan, 250012, China
| | - Gary Wong
- Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.,Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Québec, G1V 4G2, Canada
| | - Peilu Sun
- Institute of Materia Medical, Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Xuexing Zheng
- Department of Virology, School of Public Health, Shandong University, Jinan, 250012, China.
| | - Xianzhu Xia
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China
| |
Collapse
|
9
|
Li P, Wang J, Chen G, Zhang X, Lin D, Zhou Y, Yu Y, Liu W, Zhang D. Oncolytic activity of canine distemper virus in canine mammary tubular adenocarcinoma cells. Vet Comp Oncol 2019; 17:174-183. [PMID: 30756476 DOI: 10.1111/vco.12466] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/31/2019] [Accepted: 02/06/2019] [Indexed: 12/24/2022]
Abstract
Canine distemper virus (CDV), bearing a close resemblance to measles virus, represents a promising candidate for oncolytic therapy; however, its application and underlying oncolytic mechanisms in canine mammary carcinoma cells remain to be explored. Here, we found that an attenuated canine distemper vaccine strain, CDV-L, efficiently infected and inhibited the growth of canine mammary tubular adenocarcinoma CIPp cells but not MDCK cells in vitro. Transcriptomic analysis of CDV-L-infected CIPp cells revealed substantially differentially expressed genes in apoptotic and NF-κB signalling pathways. Subsequent validations confirmed that CDV-L-induced apoptosis of CIPp cells through the caspase-8 and caspase-3 pathway. Identification of phosphorylated-IκBα, phosphorylated-p65 and the nuclear translocation of p65 confirmed the activation of the NF-κB signalling pathway. Inhibition of the NF-κB pathway abrogated CDV-L-induced cleaved-caspase-3 and cleaved-PARP. In a CIPp subcutaneous xenograft mouse model, intratumoural injections of CDV-L significantly restricted tumour growth without apparent pathology, and virus remained localized within the tumour. Taken altogether, these findings indicate that CDV-L exerts an antitumour effect in CIPp cells, and that apoptosis and the NF-κB pathway play essential roles in this process.
Collapse
Affiliation(s)
- Peiran Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Jigui Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Gaoxiang Chen
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Xiaomei Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Degui Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, P.R. China
| | - Yun Zhou
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, P.R. China
| | - Yongle Yu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Weiquan Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Di Zhang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, P.R. China
| |
Collapse
|
10
|
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
|
11
|
Wang W, Xu B, Li Q, Jiang D, Yan S. Anti‑cancer effects of a novel Pan‑RAF inhibitor in a hepatocellular carcinoma cell line. Mol Med Rep 2018; 17:6185-6193. [PMID: 29484394 DOI: 10.3892/mmr.2018.8615] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 02/01/2018] [Indexed: 11/06/2022] Open
Abstract
The RAF/mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK (RAF/MEK/ERK) signaling cascade serves a prominent role in hepatocellular carcinoma (HCC) proliferation. Sorafenib (BAY 43‑9006) is a potent multikinase inhibitor of RAF kinases and a few receptor tyrosine kinases. Additionally, sorafenib causes apoptosis in a number of human tumor cell lines such as leukemia cell lines. Sorafenib is the first targeted drug to prolong the overall survival of patients with advanced HCC. However, sorafenib activity is less favorable in certain cancers, including sarcomas and melanomas, due to patient insensitivity and drug resistance. In the present study, a novel bi‑aryl urea, N‑(3‑trifluoromethylphenyl)‑N'-(2-methyl-4-(6‑cyclopropanecarboxamido-pyrimidin-4-yl) oxyphenyl) urea (CBI‑5725), is shown to be a potential candidate for the treatment of liver cancer. In the present study, the in vitro activities of CBI‑5725 and sorafenib in PLC/PRF/5 HCC cells were examined and the corresponding in vivo antitumor activities in PLC/PRF/5 human tumor xenografts. An alamar blue assay confirmed that CBI‑5725 was more cytotoxic than sorafenib to PLC/PRF/5 cells, suggesting that CBI‑5725 inhibited tumor cell proliferation more potently than sorafenib. CBI‑5725 inhibited the RAF/MEK/ERK signaling pathway to the same extent as sorafenib. In addition, CBI‑5725 elicited cell cycle arrest in the G2/M phase, while sorafenib did not markedly alter the cell cycle. Furthermore, CBI‑5725 induced apoptosis more strongly than sorafenib in a dose‑dependent manner, which may be attributed to greater caspase‑3 and poly(adenosine 5'‑diphosphate‑ribose) polymerase activation by CBI‑5725. In the PLC/PRF/5 xenograft model, 2 mg/kg CBI‑5725 inhibited tumor growth by 73%. At doses ranging from 6 to 18 mg/kg, CBI‑5725 nearly completely prevented tumor growth. These results imply that the antitumor efficacy of CBI‑5725 in HCC models may result from the suppression of the RAF/MEK/ERK signaling pathway, the induction of cell cycle arrest in the G2/M phase, and the initiation of caspase‑3‑dependent apoptosis. These observations suggested that CBI‑5725 may be a potent novel compound for the treatment of HCC.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Bo Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Qixiang Li
- Biomarker and Diagnostic Technology, Crown Bioscience Inc., Beijing 102200, P.R. China
| | - Dechun Jiang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Suying Yan
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| |
Collapse
|
12
|
Pfeffermann K, Dörr M, Zirkel F, von Messling V. Morbillivirus Pathogenesis and Virus-Host Interactions. Adv Virus Res 2018; 100:75-98. [PMID: 29551144 DOI: 10.1016/bs.aivir.2017.12.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite the availability of safe and effective vaccines against measles and several animal morbilliviruses, they continue to cause regular outbreaks and epidemics in susceptible populations. Morbilliviruses are highly contagious and share a similar pathogenesis in their respective hosts. This review provides an overview of morbillivirus history and the general replication cycle and recapitulates Morbillivirus pathogenesis focusing on common and unique aspects seen in different hosts. It also summarizes the state of knowledge regarding virus-host interactions on the cellular level with an emphasis on viral interference with innate immune response activation, and highlights remaining knowledge gaps.
Collapse
|
13
|
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
|
14
|
Pillet S, Racine T, Nfon C, Di Lenardo TZ, Babiuk S, Ward BJ, Kobinger GP, Landry N. Plant-derived H7 VLP vaccine elicits protective immune response against H7N9 influenza virus in mice and ferrets. Vaccine 2015; 33:6282-9. [PMID: 26432915 DOI: 10.1016/j.vaccine.2015.09.065] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/18/2015] [Accepted: 09/19/2015] [Indexed: 12/23/2022]
Abstract
In March 2013, the Chinese Centre for Disease Control and Prevention confirmed the first reported case of human infection with an avian influenza A H7N9 virus. Infection with this virus often caused severe pneumonia and acute respiratory distress syndrome resulting in a case fatality rate >35%. The risk of pandemic highlighted, once again, the need for a more rapid and scalable vaccine response capability. Here, we describe the rapid (19 days) development of a plant-derived VLP vaccine based on the hemagglutinin sequence of influenza H7N9 A/Hangzhou/1/2013. The immunogenicity of the H7 VLP vaccine was assessed in mice and ferrets after one or two intramuscular dose(s) with and without adjuvant (alum or GLA-SE™). In ferrets, we also measured H7-specific cell-mediated immunity. The mice and ferrets were then challenged with H7N9 A/Anhui/1/2013 influenza virus. A single immunization with the adjuvanted vaccine elicited a strong humoral response and protected mice against an otherwise lethal challenge. Two doses of unadjuvanted vaccine significantly increased humoral response and resulted in 100% protection with significant reduction of clinical signs leading to nearly asymptomatic infections. In ferrets, a single immunization with the alum-adjuvanted H7 VLP vaccine induced strong humoral and CMI responses with antigen-specific activation of CD3(+) T cells. Compared to animals injected with placebo, ferrets vaccinated with alum-adjuvanted vaccine displayed no weight loss during the challenge. Moreover, the vaccination significantly reduced the viral load in lungs and nasal washes 3 days after the infection. This candidate plant-made H7 vaccine therefore induced protective responses after either one adjuvanted or two unadjuvanted doses. Studies are currently ongoing to better characterize the immune response elicited by the plant-derived VLP vaccines. Regardless, these data are very promising for the rapid production of an immunogenic and protective vaccine against this potentially pandemic virus.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Viral/blood
- Body Weight
- Disease Models, Animal
- Female
- Ferrets
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Immunization Schedule
- Influenza A Virus, H7N9 Subtype/genetics
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza Vaccines/isolation & purification
- Injections, Intramuscular
- Lung/virology
- Male
- Mice, Inbred BALB C
- Nasal Cavity/virology
- Orthomyxoviridae Infections/pathology
- Orthomyxoviridae Infections/prevention & control
- Placebos/administration & dosage
- Plants, Genetically Modified
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Survival Analysis
- Nicotiana
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/isolation & purification
- Viral Load
Collapse
Affiliation(s)
- S Pillet
- Medicago Inc., 1020 Route de l'Église, Bureau 600, Québec, QC, Canada; Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - T Racine
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - C Nfon
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - T Z Di Lenardo
- Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - S Babiuk
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada; Department of Immunology, University of Manitoba, MB, Canada
| | - B J Ward
- Medicago Inc., 1020 Route de l'Église, Bureau 600, Québec, QC, Canada
| | - G P Kobinger
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Immunology, University of Manitoba, MB, Canada; Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - N Landry
- Medicago Inc., 1020 Route de l'Église, Bureau 600, Québec, QC, Canada.
| |
Collapse
|
15
|
Garcia JA, Ferreira HL, Vieira FV, Gameiro R, Andrade AL, Eugênio FR, Flores EF, Cardoso TC. Tumour necrosis factor-alpha-induced protein 8 (TNFAIP8) expression associated with cell survival and death in cancer cell lines infected with canine distemper virus. Vet Comp Oncol 2015; 15:336-344. [PMID: 26373887 DOI: 10.1111/vco.12168] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/17/2015] [Accepted: 07/29/2015] [Indexed: 02/06/2023]
Abstract
Oncolytic virotherapy is a novel strategy for treatment of cancer in humans and companion animals as well. Canine distemper virus (CDV), a paramyxovirus, has proven to be oncolytic through induction of apoptosis in canine-derived tumour cells, yet the mechanism behind this inhibitory action is poorly understood. In this study, three human mammary tumour cell lines and one canine-derived adenofibrosarcoma cell line were tested regarding to their susceptibility to CDV infection, cell proliferation, apoptosis, mitochondrial membrane potential and expression of tumour necrosis factor-alpha-induced protein 8 (TNFAIP8). CDV replication-induced cytopathic effect, decrease of cell proliferation rates, and >45% of infected cells were considered death and/or under late apoptosis/necrosis. TNFAIP8 and CDVM gene expression were positively correlated in all cell lines. In addition, mitochondrial membrane depolarization was associated with increase in virus titres (p < 0.005). Thus, these results strongly suggest that both human and canine mammary tumour cells are potential candidates for studies concerning CDV-induced cancer therapy.
Collapse
Affiliation(s)
- J A Garcia
- Veterinary Medicine School, Department of Support, Production and Animal Health, University of São Paulo State, Laboratory of Animal Virology and Cell Culture, Araçatuba, São Paulo, Brazil
| | - H L Ferreira
- FZEA-USP, Department of de Veterinary Medicine, Pirassununga, São Paulo, Brazil
| | - F V Vieira
- Veterinary Medicine School, Department of Support, Production and Animal Health, University of São Paulo State, Laboratory of Animal Virology and Cell Culture, Araçatuba, São Paulo, Brazil.,Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, São Paulo, Brazil
| | - R Gameiro
- Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, São Paulo, Brazil
| | - A L Andrade
- Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, São Paulo, Brazil
| | - F R Eugênio
- Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, São Paulo, Brazil
| | - E F Flores
- Department of Preventive Veterinary Medicine, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - T C Cardoso
- Veterinary Medicine School, Department of Support, Production and Animal Health, University of São Paulo State, Laboratory of Animal Virology and Cell Culture, Araçatuba, São Paulo, Brazil.,Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, São Paulo, Brazil
| |
Collapse
|
16
|
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
|
17
|
Dog nectin-4 is an epithelial cell receptor for canine distemper virus that facilitates virus entry and syncytia formation. Virology 2012; 436:210-20. [PMID: 23260107 DOI: 10.1016/j.virol.2012.11.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2012] [Revised: 10/08/2012] [Accepted: 11/19/2012] [Indexed: 11/22/2022]
Abstract
Canine distemper virus (CDV) was shown to use dog nectin-4 as a receptor to gain entry into epithelial cells. RNA from dog placenta or MDCK kidney cells was isolated and cDNAs were prepared. Two splice variants of dog nectin-4 were identified. A deletion of 25 amino acids was found in the cytoplasmic domain of dog nectin-4 from MDCK cells, corresponding to a splice variant that is also seen in murine nectin-4, and did not affect its role as a receptor. Both dog nectin-4 and human nectin-4 could function as an entry factor for CDV containing an EGFP reporter gene. Inhibition of dog nectin-4 expression by RNAi or nectin-4 antibodies decreased CDV titers and EGFP fluorescence. Finally, dog nectin-4 also promotes syncytia formation, which could be inhibited by siRNA treatment. These data confirm that dog nectin-4 can be used by CDV to gain entry into epithelial cells, and facilitate virus spread.
Collapse
|
18
|
Noyce RS, Richardson CD. Nectin 4 is the epithelial cell receptor for measles virus. Trends Microbiol 2012; 20:429-39. [PMID: 22721863 DOI: 10.1016/j.tim.2012.05.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 05/14/2012] [Accepted: 05/23/2012] [Indexed: 01/06/2023]
Abstract
Measles virus (MV) causes acute respiratory disease, infects lymphocytes and multiple organs, and produces immune suppression leading to secondary infections. In rare instances it can also cause persistent infections in the brain and central nervous system. Vaccine and laboratory-adapted strains of MV use CD46 as a receptor, whereas wild-type strains of MV (wtMV) cannot. Both vaccine and wtMV strains infect lymphocytes, monocytes, and dendritic cells (DCs) using the signaling lymphocyte activation molecule (CD150/SLAM). In addition, MV can infect the airway epithelial cells of the host. Nectin 4 (PVRL4) was recently identified as the epithelial cell receptor for MV. Coupled with recent observations made in MV-infected macaques, this discovery has led to a new paradigm for how the virus accesses the respiratory tract and exits the host. Nectin 4 is also a tumor cell marker which is highly expressed on the apical surface of many adenocarcinoma cell lines, making it a potential target for MV oncolytic therapy.
Collapse
Affiliation(s)
- Ryan S Noyce
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | | |
Collapse
|