1
|
Lu X, Wang X, Liu X, Liu X. The multifaceted interactions between Newcastle disease virus proteins and host proteins: a systematic review. Virulence 2024; 15:2299182. [PMID: 38193514 PMCID: PMC10793697 DOI: 10.1080/21505594.2023.2299182] [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: 10/06/2023] [Accepted: 12/20/2023] [Indexed: 01/10/2024] Open
Abstract
Newcastle disease virus (NDV) typically induces severe illness in poultry and results in significant economic losses for the worldwide poultry sector. NDV, an RNA virus with a single-stranded negative-sense genome, is susceptible to mutation and immune evasion during viral transmission, thus imposing enormous challenges to avian health and poultry production. NDV is composed of six structural proteins and two nonstructural proteins that exert pivotal roles in viral infection and antiviral responses by interacting with host proteins. Nowadays, there is a particular focus on the mechanisms of virus-host protein interactions in NDV research, yet a comprehensive overview of such research is still lacking. Herein, we briefly summarize the mechanisms regarding the effects of virus-host protein interaction on viral infection, pathogenesis, and host immune responses. This review can not only enhance the present comprehension of the mechanism underlying NDV and host interplay, but also furnish a point of reference for the advancement of antiviral measures.
Collapse
Affiliation(s)
- Xiaolong Lu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaowen Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
2
|
Zhu H, Hou P, Chu F, Li X, Zhang W, Sun X, Liu Y, Zhao G, Gao Y, He DC, Wang H, He H. PBLD promotes IRF3 mediated the type I interferon (IFN-I) response and apoptosis to inhibit viral replication. Cell Death Dis 2024; 15:727. [PMID: 39362857 PMCID: PMC11450232 DOI: 10.1038/s41419-024-07083-w] [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: 01/19/2024] [Revised: 09/07/2024] [Accepted: 09/16/2024] [Indexed: 10/05/2024]
Abstract
Recent studies have implicated the phenazine biosynthesis-like domain-containing protein (PBLD) in the negative regulation of the development and progression of various cancers. However, its function in viral infection remains unknown. In this study, we found that PBLD plays important roles in multiple virus infections including BPIV3, SeV, VSV, and HSV-1. Our study revealed that PBLD enhances the expression of type I interferon (IFN-I) and ISGs through interferon regulatory factor 3 (IRF3). Further study indicated that PBLD promotes transcriptional phosphorylation of IRF3 (S385/386), thereby facilitating virus-induced IFN-I production. Interestingly, PBLD mediates virus-triggered mitochondrial apoptosis through its dependence on IRF3 (K313/315). Mechanistically, PBLD facilitated virus-induced apoptosis by recruiting the Puma protein to the mitochondria via IRF3. Additionally, we performed mutational analyses of IRF3, showing that its loss of either transcriptional or apoptotic function markedly increased viral replication. Moreover, macrophages with PBLD deficiency during viral infection exhibited decreased the IFN-I and ISGs expression, exacerbating viral infection. Importantly, mice deficient in PBLD exhibited increased viral replication and susceptibility to SeV infection, leading to decreased survival. Notably, Cedrelone, a chemical activator of PBLD, has the ability to reduce SeV replication. Collectively, we first discovered the new function of PBLD in viral infection, broadening our understanding of potential therapeutic targets and offering new insights for antiviral drug development.
Collapse
Affiliation(s)
- Hongchao Zhu
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Peili Hou
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Fengyun Chu
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Xingyu Li
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Wenjia Zhang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Xiaonan Sun
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Yu Liu
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Guimin Zhao
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Yuwei Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Daniel Chang He
- The College of Arts and Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hongmei Wang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China.
| | - Hongbin He
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China.
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China.
| |
Collapse
|
3
|
Li D, Mo R, Li X, Cheng R, Xie J, Li H, Yang Y, Li S, Li H, Yan Z, Wei S, Idris A, Li X, Feng R. Mammalian orthoreovirus capsid protein σ3 antagonizes RLR-mediated antiviral responses by degrading MAVS. mSphere 2024; 9:e0023624. [PMID: 38757961 PMCID: PMC11332348 DOI: 10.1128/msphere.00236-24] [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: 03/21/2024] [Accepted: 04/02/2024] [Indexed: 05/18/2024] Open
Abstract
Mammalian orthoreovirus (MRV) outer capsid protein σ3 is a multifunctional protein containing a double-stranded RNA-binding domain, which facilitates viral entry and assembly. We reasoned that σ3 has an innate immune evasion function. Here, we show that σ3 protein localizes in the mitochondria and interacts with mitochondrial antiviral signaling protein (MAVS) to activate the intrinsic mitochondria-mediated apoptotic pathway. Consequently, σ3 protein promotes the degradation of MAVS through the intrinsic caspase-9/caspase-3 apoptotic pathway. Moreover, σ3 protein can also inhibit the expression of the components of the RNA-sensing retinoic acid-inducible gene (RIG)-like receptor (RLR) signaling pathway to block antiviral type I interferon responses. Mechanistically, σ3 inhibits RIG-I and melanoma differentiation-associated gene 5 expression is independent of its inhibitory effect on MAVS. Overall, we demonstrate that the MRV σ3 protein plays a vital role in negatively regulating the RLR signaling pathway to inhibit antiviral responses. This enables MRV to evade host defenses to facilitate its own replication providing a target for the development of effective antiviral drugs against MRV. IMPORTANCE Mammalian orthoreovirus (MRV) is an important zoonotic pathogen, but the regulatory role of its viral proteins in retinoic acid-inducible gene-like receptor (RLR)-mediated antiviral responses is still poorly understood. Herein, we show that MRV σ3 protein co-localizes with mitochondrial antiviral signaling protein (MAVS) in the mitochondria and promotes the mitochondria-mediated intrinsic apoptotic pathway to cleave and consequently degrade MAVS. Furthermore, tryptophan at position 133 of σ3 protein plays a key role in the degradation of MAVS. Importantly, we show that MRV outer capsid protein σ3 is a key factor in antagonizing RLR-mediated antiviral responses, providing evidence to better unravel the infection and transmission mechanisms of MRV.
Collapse
Affiliation(s)
- Dianyu Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Rongqian Mo
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Xiaoyi Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Rongrong Cheng
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Jingying Xie
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Hongshan Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Yanmei Yang
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Shasha Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Huixia Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Zhenfang Yan
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Suocheng Wei
- College of Life science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Adi Idris
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Xiangrong Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Ruofei Feng
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| |
Collapse
|
4
|
Zheng L, Li Z, Wang R, Wang J, Liu B, Wang Y, Qin S, Yang J, Liu J. A novel photosensitizer DTPP-mediated photodynamic therapy induces oxidative stress and apoptosis through mitochondrial pathways in LA795 cells. Photodiagnosis Photodyn Ther 2024; 45:103894. [PMID: 37984526 DOI: 10.1016/j.pdpdt.2023.103894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
OBJECTIVE Investigation of the effects of 5-5- (4-N, N-diacetoxylphenyl)-10,15,20- tetraphenylporphyrin (DTPP)-mediated photodynamic therapy (PDT) on oxidative stress and mitochondrial apoptosis in LA795 lung cancer cells. METHODS Proteomics was used to identify differentially expressed proteins after PDT treatment. The apoptosis rate was determined by flow cytometry. Morphologic observation of apoptosis, reactive oxygen species (ROS) levels, antioxidant indices, nitric oxide (NO) content, mitochondrial membrane potential (MMP), and Caspase- 9 and Caspase-3 were determined by assays; apoptosis-related protein levels of Cytochrome (Cyto) c, Bcl- 2, Bax were determined by Western blot. RESULTS Typical apoptosis morphology of LA795 cells was observed after PDT. The cells were mainly in the apoptosis death pathway with high cell apoptosis rates. The proteomics study observed the apoptosis-associated proteins, oxidative stress proteins, antioxidant proteins, the cytoskeletal protein and mitochondrial dysfunction in LA 795 cells. Additional results indicated that PDT could increase levels of ROS, NO; decrease glutathione (GSH) content and MMP; upregulated Bax, Cyto c, and Caspase-3 protein expression, inhibited Bcl-2 protein expression, and further induced cell apoptosis. The effect of DTPP-PDT on lung cancer was: first, mitochondrial Cyto c is released into the cytoplasm, then Caspase- 9 / Caspase-3 was activated, Bcl-2 decreased/Bax increased, initiating cell apoptosis. CONCLUSION DTPP-PDT could induce oxidative stress and apoptosis via mitochondrial pathways in LA795 cells.
Collapse
Affiliation(s)
- Liqing Zheng
- Hebei Key Laboratory of Neuropharmacology, Department of Pharmacy, Hebei North University, Zhangjiakou 075000, China
| | - Ze Li
- Hebei Key Laboratory of Neuropharmacology, Department of Pharmacy, Hebei North University, Zhangjiakou 075000, China
| | - Ruibo Wang
- Hebei Key Laboratory of Neuropharmacology, Department of Pharmacy, Hebei North University, Zhangjiakou 075000, China
| | - Jing Wang
- Hebei North University Library Zhangjiakou 075000, China
| | - Bochao Liu
- Tianjin Shuangling Middle School, Tianjin 300041 China
| | - Yiying Wang
- Hebei Key Laboratory of Neuropharmacology, Department of Pharmacy, Hebei North University, Zhangjiakou 075000, China
| | - Shihao Qin
- Hebei Key Laboratory of Neuropharmacology, Department of Pharmacy, Hebei North University, Zhangjiakou 075000, China
| | - Junying Yang
- Hebei Key Laboratory of Neuropharmacology, Department of Pharmacy, Hebei North University, Zhangjiakou 075000, China
| | - Jianhua Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou 075000, China.
| |
Collapse
|
5
|
Li S, Xu B, Luo Y, Luo J, Huang S, Guo X. Autophagy and Apoptosis in Rabies Virus Replication. Cells 2024; 13:183. [PMID: 38247875 PMCID: PMC10814280 DOI: 10.3390/cells13020183] [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: 11/14/2023] [Revised: 12/28/2023] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
Rabies virus (RABV) is a single-stranded negative-sense RNA virus belonging to the Rhabdoviridae family and Lyssavirus genus, which is highly neurotropic and can infect almost all warm-blooded animals, including humans. Autophagy and apoptosis are two evolutionarily conserved and genetically regulated processes that maintain cellular and organismal homeostasis, respectively. Autophagy recycles unnecessary or dysfunctional intracellular organelles and molecules in a cell, whereas apoptosis eliminates damaged or unwanted cells in an organism. Studies have shown that RABV can induce both autophagy and apoptosis in target cells. To advance our understanding of pathogenesis of rabies, this paper reviews the molecular mechanisms of autophagy and apoptosis induced by RABV and the effects of the two cellular events on RABV replication.
Collapse
Affiliation(s)
- Saisai Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (S.L.); (Y.L.)
| | - Bowen Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China;
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (S.L.); (Y.L.)
| | - Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (S.L.); (Y.L.)
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA;
- Department of Hematology and Oncology, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (S.L.); (Y.L.)
| |
Collapse
|
6
|
Adam FEA, Zhao X, Guan Z, Chang Z, Thrusfield M, Lu K, El Tigani-Asil ETA, Terab AMA, Ismael M, Tong L, Prince-Theodore DW, Luo C, Xiao S, Wang X, Liu H, Yang Z. Simultaneous Expression of Chicken Granulocyte Monocyte Colony-Stimulating Factor and the Hemagglutinin-Neuraminidase Epitope of the Virulent Newcastle Disease Virus Genotype VII C22 Strain in a Functional Synthetic Recombinant Adenovirus as a Genotype-Matched Vaccine with Potential Antiviral Activity. Microbiol Spectr 2023; 11:e0402422. [PMID: 37036344 PMCID: PMC10269747 DOI: 10.1128/spectrum.04024-22] [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: 10/04/2022] [Accepted: 03/18/2023] [Indexed: 04/11/2023] Open
Abstract
When it comes to the prevention of clinical signs and mortality associated with infection of the Newcastle disease virus (NDV), vaccination has been very effective. However, recent evidence has proven that more highly virulent strains are emerging that bypass existing immune protection and pose a serious threat to the global poultry industry. Here, a novel rescued adenovirus 5-coexpressed chicken granulocyte monocyte colony-stimulating factor (ChGM-CSF) bio-adjuvant and C22-hemagglutinin-neuraminidase (HN) boosted chickens' immunological genetic resistance and thus improved the immunological effectiveness of the critical new-generation vaccine in vitro and in vivo. Accordingly, the hemagglutination inhibition (HI) titers (log2) of the recombinant adenovirus (rAdv)-ChGM-CSF-HN-immunized chickens had greater, more persistent, and longer-lasting NDV-specific antibodies than the La Sota and rAdv-HN-inoculated birds. Moreover, humoral and adaptive immunological conditions were shown to be in harmony after rAdv-ChGM-CSF-HN inoculation and uniformly enhanced the expression of alpha interferon (IFN-α), IFN-β, IFN-γ, interleukin-1β (IL-1β), IL-2, IL-16, IL-18, and IL-22. Postchallenge, the control challenge (CC), wild-type adenovirus (wtAdv), and rAdv-ChGM-CSF groups developed unique NDV clinical manifestations, significant viral shedding, high tissue viral loads, gross and microscopic lesions, and 100% mortality within 7 days. The La Sota, rAdv-HN, and rAdv-ChGM-CSF-HN groups were healthy and had 100% survival rates. The rAdv-ChGM-CSF-HN group swiftly regulated and stopped viral shedding and had lower tissue viral loads than all groups at 5 days postchallenge (dpc). Thus, the antiviral activity of ChGM-CSF offered robust immune protection in the face of challenge and reduced viral replication convincingly. Our advance innovation concepts, combining ChGM-CSF with a field-circulating strain epitope, could lead to the development of a safe, genotype-matched, universal transgenic vaccine that could eradicate the disease globally, reducing poverty and food insecurity. IMPORTANCE We studied the biological characterization of the developed functional synthetic recombinant adenoviruses, which showed a high degree of safety, thermostability, and genetic stability for up to 20 passages. It was demonstrated through both in vitro and in vivo testing that the immunogenicity of the proposed vaccine, which uses the T2A peptide from the Thosea asigna virus capsid protein supported by glycine and serine, helps with efficiency to generate a multicistronic vector, enables expression of two functional proteins in rAdv-ChGM-CSF-HN, and is superior to that of comparable vaccines. Additionally, adenovirus can be used to produce vaccines matching the virulent field-circulating strain epitope. Because there is no preexisting human adenoviral immunity detected in animals, the potency of adenoviral vaccines looks promising. Also, it ensures that the living vector does not carry the resistance gene that codes for the kanamycin antibiotic. Accordingly, a human recombinant adenoviral vaccine that has undergone biological improvements is beneficial and important.
Collapse
Affiliation(s)
- Fathalrhman Eisa Addoma Adam
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Department of Preventive Medicine and Public Health, Faculty of Veterinary Science, University of Nyala, Nyala, Sudan
| | - Xueliang Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Zhao Guan
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Zhengwu Chang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Michael Thrusfield
- Veterinary Clinical Sciences Royal (Dick) School of Veterinary Studies, University of Edinburgh, Roslin, Midlothian, United Kingdom
| | - Kejia Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - El Tigani Ahmed El Tigani-Asil
- Veterinary Laboratories Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi, United Arab Emirates
| | - Abdelnasir Mohammed Adam Terab
- Veterinary Laboratories Division, Animal Wealth Sector, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi, United Arab Emirates
| | - Mohamedelfateh Ismael
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Lina Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | | | - Chen Luo
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Sa Xiao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xinglong Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Haijin Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
7
|
Nayak BN, Rajagopal K, Shunmugasundaram R, Rao PL, Vaidyanathan S, Subbiah M. Molecular characterization suggests kinetic modulation of expression of accessory viral protein, W, in Newcastle disease virus infected DF1 cells. Virusdisease 2023; 34:236-247. [PMID: 37408548 PMCID: PMC10317930 DOI: 10.1007/s13337-023-00813-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 03/01/2023] [Indexed: 07/07/2023] Open
Abstract
Viruses adopt strategies to efficiently utilize their compact genome. Members of the family Paramyxoviridae, exhibit a cotranscriptional RNA editing mechanism wherein polymerase stuttering generates accessory proteins from Phosphoprotein (P) gene. Newcastle disease virus (NDV), an avian paramyxovirus, expresses two accessory proteins, V and W, by RNA editing. While P and V proteins are well studied, very little is known about W protein. Recent studies confirmed W protein expression in NDV and the unique subcellular localization of W proteins of virulent and avirulent NDV. We characterized the W protein of NDV strain Komarov, a moderately virulent vaccine strain. W mRNA expression ranged between 7 and 9% of total P gene transcripts similar to virulent NDV. However, W protein expression, detectable by 6 h, peaked at 24 h and dropped by 48 h post infection in DF1 cells indicating a kinetically regulated expression by the virus. The W protein localized in the nucleus and by mutations, a strong nuclear localization signal was identified in the C-terminal region of W protein. The viral growth kinetics study suggested neither supplementation of W protein nor subcellular localization pattern of the supplemented W protein influenced viral replication in vitro similar to that noticed in avirulent NDV. A cytoplasmic mutant of W protein localized in cytoplasm unlike specific mitochondrial colocalization as recorded in velogenic NDV strain SG10 indicating a possible role of W protein in determining the viral pathogenicity. This study describes for the first time, the distinct features of W protein of moderately virulent NDV. Supplementary Information The online version contains supplementary material available at 10.1007/s13337-023-00813-2.
Collapse
Affiliation(s)
- B. Nagaraj Nayak
- National Institute of Animal Biotechnology, Hyderabad, Telangana India
- Regional Centre for Biotechnology, New Delhi, India
| | | | | | | | | | - Madhuri Subbiah
- National Institute of Animal Biotechnology, Hyderabad, Telangana India
- Regional Centre for Biotechnology, New Delhi, India
| |
Collapse
|
8
|
Chu Z, Yang S, Li Q, Shang J, Ren Z, Ren F. The V protein in oncolytic Newcastle disease virus promotes HepG2 hepatoma cell proliferation at the single-cell level. BMC Cancer 2023; 23:346. [PMID: 37069523 PMCID: PMC10108501 DOI: 10.1186/s12885-023-10815-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 04/06/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Newcastle disease virus (NDV) is an oncolytic virus that can inhibit cancer cell proliferation and kill cancer cells. The NDV nonstructural V protein can regulate viral replication; however, whether the V protein contributes to NDV oncolysis is unclear. RESULTS This study revealed that NDV inhibited tumor cell proliferation and that V protein expression promoted the proliferation of HepG2 cells, as determined at the single-cell level. In addition, to identify the regulatory mechanism of the V protein in HepG2 cells, transcriptome sequencing was performed and indicated that the expression/activation of multiple cell proliferation-related genes/signaling pathways were changed in cells overexpressing the V protein. Hence, the MAPK and WNT signaling pathways were selected for verification, and after blocking these two signaling pathways with inhibitors, the V protein promotion of cell proliferation was found to be attenuated. CONCLUSIONS The results showed that the V protein regulated the proliferation of cancer cells through multiple signaling pathways, providing valuable references for future studies on the mechanism by which the V protein regulates cancer cell proliferation.
Collapse
Affiliation(s)
- Zhili Chu
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
- Henan International Joint Laboratory of Immunity and Targeted Therapy for liver-intestinal Tumors, Xinxiang Medical University, Xinxiang, China.
| | - Sihui Yang
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Qianru Li
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jianing Shang
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Zilong Ren
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Feng Ren
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
- Henan International Joint Laboratory of Immunity and Targeted Therapy for liver-intestinal Tumors, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
9
|
Leśniak W, Bohush A, Maksymowicz M, Piwowarczyk C, Karolak NK, Jurewicz E, Filipek A. Involvement of CacyBP/SIP in differentiation and the immune response of HaCaT keratinocytes. Immunobiology 2023; 228:152385. [PMID: 37156124 DOI: 10.1016/j.imbio.2023.152385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/05/2023] [Accepted: 04/10/2023] [Indexed: 05/10/2023]
Abstract
CacyBP/SIP is a multifunctional protein present in various cells and tissues. However, its expression and role in the epidermis has not been explored so far. In this work, using RT-qPCR, Western blot analysis and three-dimensional (3D) organotypic cultures of HaCaT keratinocytes we show that CacyBP/SIP is present in the epidermis. To investigate the possible role of CacyBP/SIP in keratinocytes we obtained CacyBP/SIP knockdown cells and studied the effect of CacyBP/SIP deficiency on their differentiation and response to viral infection. We found that CacyBP/SIP knockdown results in reduced expression of epidermal differentiation markers in both undifferentiated and differentiated HaCaT cells. Since epidermis is engaged in immune defense, the impact of CacyBP/SIP knockdown on this process was also analyzed. By applying RT-qPCR and Western blot it was found that poly(I:C), a synthetic analog of double-stranded RNA that mimics viral infection, stimulated the expression of genes involved in antiviral response, such as IFIT1, IFIT2 and OASL. Interestingly, following poly(I:C) stimulation, the level of expression of these genes was significantly lower in cells with CacyBP/SIP knockdown than control ones. Since the signaling pathway mediating cellular responses to viral infection involves, among others, the STAT1 transcription factor, we measured its activity using luciferase assay and found that it was lower in CacyBP/SIP knockdown HaCaT cells. Altogether, the presented results indicate that CacyBP/SIP promotes epidermal differentiation and might be involved in response of the skin cells to viral infection.
Collapse
Affiliation(s)
- Wiesława Leśniak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland.
| | - Anastasiia Bohush
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Małgorzata Maksymowicz
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Cezary Piwowarczyk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Natalia Katarzyna Karolak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; Department of Chemistry, University of Warsaw, 1 Pasteur Street, 02-093 Warsaw, Poland
| | - Ewelina Jurewicz
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Anna Filipek
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland.
| |
Collapse
|
10
|
Zhang D, Ding Z, Xu X. Pathologic Mechanisms of the Newcastle Disease Virus. Viruses 2023; 15:v15040864. [PMID: 37112843 PMCID: PMC10143668 DOI: 10.3390/v15040864] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/18/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023] Open
Abstract
Newcastle disease (ND) has been a consistent risk factor to the poultry industry worldwide. Its pathogen, Newcastle disease virus (NDV), is also a promising antitumor treatment candidate. The pathogenic mechanism has intrigued the great curiosity of researchers, and advances in the last two decades have been summarized in this paper. The NDV’s pathogenic ability is highly related to the basic protein structure of the virus, which is described in the Introduction of this review. The overall clinical signs and recent findings pertaining to NDV-related lymph tissue damage are then described. Given the involvement of cytokines in the overall virulence of NDV, cytokines, particularly IL6 and IFN expressed during infection, are reviewed. On the other hand, the host also has its way of antagonizing the virus, which starts with the detection of the pathogen. Thus, advances in NDV’s physiological cell mechanism and the subsequent IFN response, autophagy, and apoptosis are summarized to provide a whole picture of the NDV infection process.
Collapse
|
11
|
Abstract
Avian paramyxovirus 1 (APMV-1), also known as Newcastle disease virus (NDV), causes severe and economically important disease in poultry around the globe. Although a limited amount of APMV-1 strains in urban areas have been characterized, the role of the urban wild bird population as an APMV-1 reservoir is unclear. Because urban birds may have an important role for long-term circulation of the virus, fecal and swab samples were collected by community scientists from wild birds in New York City (NYC), New York, United States. These samples were screened for APMV-1 and genotypically characterized by sequencing of the complete genome. A total of 885 samples were collected from NYC parks and from a local wildlife rehabilitation clinic from October 2020 through June 2021, and 255 samples obtained from 197 birds have been processed to date. Eight birds (4.1%) screened positive for the APMV-1 nucleoprotein gene by conventional reverse transcription PCR (RT-PCR), and two live viruses were isolated via egg culture. A multibasic F protein cleavage sequence, 112R R K K R F117, an indicator of highly pathogenic velogenic APMV-1 strains, was present in the two samples fully sequenced by next generation sequencing. Phylogenetic analysis of the F gene coding sequence classified both isolates into genotype VI, a diverse and predominant genotype responsible for APMV-1 outbreaks in pigeon and dove species worldwide. IMPORTANCE Here we describe the first large-scale effort to screen for APMV-1 in New York City’s wild bird population as part of the New York City Virus Hunters program, a community science initiative. We characterized two isolates of APMV-1, with phylogenetic analyses suggesting diversity in established and circulating strains of pigeon paramyxoviruses. Our isolates are also domestic reference strains for future APMV-1 vaccine developments. Future surveillance in this region may contribute to our understanding of APMV-1’s evolution and genetic diversity, as well as inform poultry husbandry and vaccination practices in New York State.
Collapse
|
12
|
Wang C, Wang T, Duan L, Chen H, Hu R, Wang X, Jia Y, Chu Z, Liu H, Wang X, Zhang S, Xiao S, Wang J, Dang R, Yang Z. Evasion of Host Antiviral Innate Immunity by Paramyxovirus Accessory Proteins. Front Microbiol 2022; 12:790191. [PMID: 35173691 PMCID: PMC8841848 DOI: 10.3389/fmicb.2021.790191] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/22/2021] [Indexed: 01/01/2023] Open
Abstract
For efficient replication, viruses have developed multiple strategies to evade host antiviral innate immunity. Paramyxoviruses are a large family of enveloped RNA viruses that comprises diverse human and animal pathogens which jeopardize global public health and the economy. The accessory proteins expressed from the P gene by RNA editing or overlapping open reading frames (ORFs) are major viral immune evasion factors antagonizing type I interferon (IFN-I) production and other antiviral innate immune responses. However, the antagonistic mechanisms against antiviral innate immunity by accessory proteins differ among viruses. Here, we summarize the current understandings of immune evasion mechanisms by paramyxovirus accessory proteins, specifically how accessory proteins directly or indirectly target the adaptors in the antiviral innate immune signaling pathway to facilitate virus replication. Additionally, some cellular responses, which are also involved in viral replication, will be briefly summarized.
Collapse
|
13
|
Nan FL, Zhang H, Nan WL, Xie CZ, Ha Z, Chen X, Xu XH, Qian J, Qiu XS, Ge JY, Bu ZG, Zhang Y, Lu HJ, Jin NY. Lentogenic NDV V protein inhibits IFN responses and represses cell apoptosis. Vet Microbiol 2021; 261:109181. [PMID: 34399297 DOI: 10.1016/j.vetmic.2021.109181] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 07/01/2021] [Indexed: 01/13/2023]
Abstract
The V protein of Newcastle disease virus (NDV) has been shown to inhibit the secretion of interferon (IFN) during infection, which is responsible for the promotion of NDV pathogenicity. However, the ability of the V protein to suppress host innate immunity is not well understood. In this study, we explored the function of V protein and its relationship with virulence by generating V protein-inserted recombinant (r) NDVs. Using rNDVs as a model, we examined the efficiency of infection, IFN responses, and apoptosis of host cells during infection. We found that viral propagation occurred smoothly when V protein from lentogenic NDV is inserted instead of the V protein from the velogenic strain. The infection of lentogenic V protein-inserted rNDV induced less expression of IFNs and downstream antiviral proteins via efficient degradation of p-STAT1 and MDA5. Moreover, velogenic V protein triggered a higher apoptosis rate during infection thereby restricting the replication of NDV. Conversely, lentogenic V protein inhibits IFN responses efficiently and induces less apoptosis compared to the velogenic strain. Our findings provide a novel understanding of the role of V protein in NDV pathogenicity.
Collapse
Affiliation(s)
- Fu Long Nan
- College of Veterinary Medicine, College of Animal Science, Jilin University, Changchun, 130062, China; Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, 130122, China
| | - He Zhang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Wen Long Nan
- China Animal Health and Epidemiology Center, Qingdao, 266032, China
| | - Chang Zhan Xie
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Zhuo Ha
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Xing Chen
- Changchun Institute of Biological Products Co., Ltd. Changchun, 130012, China
| | - Xiao Hong Xu
- College of Veterinary Medicine, College of Animal Science, Jilin University, Changchun, 130062, China
| | - Jing Qian
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Xu Sheng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Jin Ying Ge
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China
| | - Zhi Gao Bu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China
| | - Ying Zhang
- College of Veterinary Medicine, College of Animal Science, Jilin University, Changchun, 130062, China.
| | - Hui Jun Lu
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, 130122, China.
| | - Ning Yi Jin
- College of Veterinary Medicine, College of Animal Science, Jilin University, Changchun, 130062, China; Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, 130122, China.
| |
Collapse
|
14
|
Tong L, Chu Z, Gao X, Yang M, Adam FEA, Theodore DWP, Liu H, Wang X, Xiao S, Yang Z. Newcastle disease virus V protein interacts with hnRNP H1 to promote viral replication. Vet Microbiol 2021; 260:109093. [PMID: 34265512 DOI: 10.1016/j.vetmic.2021.109093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 04/19/2021] [Indexed: 11/17/2022]
Abstract
The interactions between host cellular proteins and viral proteins are important for successful infection by viruses. Previous studies from our group have identified various host cellular proteins that can interact with the Newcastle disease virus V protein (Chu et al., 2018a), but their function in NDV replication has not been fully determined. The present study reports that heterogenous nuclear ribonucleoprotein H1 (hnRNP H1) can interact with NDV V protein in yeast. The immunofluorescence results showed that hnRNP H1 and V protein could colocalize in the cytoplasm of a chicken embryo fibroblast cell line (DF-1 cells). Co-immunoprecipitation assays further verified the interaction of these two proteins. The effects of overexpression and knockdown of hnRNP H1 on NDV replication were evaluated in DF-1 cells through real time quantitative PCR (RT-qPCR) and plaque assays. The regulation of V protein on hnRNP H1 expression was also examined. The results indicated that overexpression of hnRNP H1 facilitated NDV replication, while knockdown of hnRNP H1 decreased NDV replication. It was also shown that V protein could regulate hnRNP H1 expression at the protein level instead of the transcription level. The effect of V protein and hnRNP H1 on the DF-1 cell cycle was also tested and the results revealed that V protein may regulate cell proliferation by controlling the expression of hnRNP H1. Taken together, these results suggest that NDV V protein could promote viral replication by interacting with hnRNP H1.
Collapse
Affiliation(s)
- Lina Tong
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, 712100, China; College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai, 810000, China
| | - Zhili Chu
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, 712100, China; School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Xiaolong Gao
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai, 810000, China
| | - Mengqing Yang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, 712100, China
| | - Fathalrhman Eisa A Adam
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, 712100, China
| | | | - Haijin Liu
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, 712100, China
| | - Xinglong Wang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, 712100, China
| | - Sa Xiao
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, 712100, China
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
15
|
Li J, Yang L, Mao L, Li W, Sun M, Liu C, Xue T, Zhang W, Liu M, Li B. Caprine parainfluenza virus type 3 N protein promotes viral replication via inducing apoptosis. Vet Microbiol 2021; 259:109129. [PMID: 34087675 DOI: 10.1016/j.vetmic.2021.109129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/16/2021] [Indexed: 11/28/2022]
Abstract
Caprine parainfluenza virus type 3 (CPIV3) is one of the most important viral respiratory pathogens of goat. Accumulating evidence demonstrates that apoptosis is a cellular mechanism for the host response to pathogens, and it participates in regulating viral replication. However, there is little study on CPIV3-induced host cells apoptosis. In this study, primary goat tracheal epithelial (GTE) cells were established as a cellular model that is permissive to CPIV3 infection. Then, we showed that CPIV3 infection induced apoptosis in GTE cells, as determined by morphological changes, flow cytometry and TUNEL assay. Moreover, Caspase activity and the expression of pro-apoptotic genes further suggested that CPIV3 induced apoptosis by activating both the intrinsic and extrinsic pathways. Mechanistically, the ability of CPIV3 to induce apoptosis was activated by N protein, and the viral protein increased CPIV3 replication through effecting apoptosis. Overall, our findings showed that GTE cells that will enable further analysis of CPIV3 infection and offers novel insights into the mechanisms of CPIV3-induced apoptosis in host cells.
Collapse
Affiliation(s)
- Jizong Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China; School of Pharmacy, Linyi University, Linyi, 276000, China; Institute of Life Sciences, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Leilei Yang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Li Mao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Wenliang Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China; Institute of Life Sciences, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Min Sun
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Chuanmin Liu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China; School of Pharmacy, Linyi University, Linyi, 276000, China; Institute of Life Sciences, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - Tao Xue
- School of Pharmacy, Linyi University, Linyi, 276000, China
| | - Wenwen Zhang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China
| | - Maojun Liu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China; Institute of Life Sciences, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Bin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, 210014, China; Institute of Life Sciences, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
16
|
Yang M, Ma J, Chu Z, Cao X, Lu K, Shi X, Tong L, Yan C, Liu H, Wang X, Xiao S, Yang Z. Musashi1 inhibit the release of Newcastle disease viruses through preventing apoptosis of DF-1 cells. Poult Sci 2021; 100:101105. [PMID: 34062443 PMCID: PMC8173301 DOI: 10.1016/j.psj.2021.101105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/19/2021] [Accepted: 02/27/2021] [Indexed: 12/22/2022] Open
Abstract
The efficient proliferation of Newcastle disease virus (NDV) depends on its inhibition of host cell innate immunity. V protein acts as a nonstructural protein which plays a significant role in virus replication, whereas its function remains to be further explored. In this study, Musashi RNA binding protein 1 (MSI1) was selected and its interaction with V protein was further verified by Co-immunoprecipitation (Co-IP) and Immuno-colocalization test. Through the transfection of pCMV-HA-MSI1 in DF-1 cells, the overexpression of MSI1 reduced virus particles in the cell supernatant but not reduced mRNA and virus protein in cells pellet, which suggests that MSI1may act as a new antiviral molecule by inhibiting viral release. Cell early apoptosis was detected by flow cytometry (FCM), the result shows that overexpression of MSI1 inhibit cell apoptosis, implying MSI1 Inhibit virus release may through this way. Taken together, MSI1 and NDV V protein has a detectable interaction, and may block apoptosis to inhibit the release of NDV. However, this is the first report about the interaction between MSI1 and V protein of NDV that can inhibit the NDV replicated.
Collapse
Affiliation(s)
- Mengqing Yang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Jiangang Ma
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Zhili Chu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Xuhong Cao
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Kejia Lu
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Xiaolei Shi
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Lina Tong
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Chuanqi Yan
- Bureau of Agriculture and Rural Affairs of Huangdao, Qingdao, Shandong 266400, China
| | - Haijin Liu
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Xinglong Wang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Sa Xiao
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
17
|
Del Vesco AP, Jang HJ, Monson MS, Lamont SJ. Role of the chicken oligoadenylate synthase-like gene during in vitro Newcastle disease virus infection. Poult Sci 2021; 100:101067. [PMID: 33752069 PMCID: PMC8005822 DOI: 10.1016/j.psj.2021.101067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/19/2021] [Accepted: 02/11/2021] [Indexed: 01/21/2023] Open
Abstract
The enzyme 2′-5′ oligoadenylate synthase (OAS) is one of the key interferon-induced antiviral factors that act through inhibition of viral replication. In chickens, there is a single well-characterized OAS gene, oligoadenylate synthase-like (OASL) that has been shown to be upregulated after infection with various viruses. However, a deeper understanding of how chicken OASL acts against viral infection is still necessary. In this study, we tested the hypothesis that OASL short interfering RNA (siRNA)–mediated knockdown would decrease the host gene expression response to the Newcastle disease virus (NDV) by impacting antiviral pathways. To assess our hypothesis, a chicken fibroblast cell line (DF-1) was infected with the NDV (LaSota strain) and OASL expression was knocked down using a specific siRNA. The level of NDV viral RNA in the cells and the expression of interferon response- and apoptosis-related genes were evaluated by quantitative PCR at 4, 8, and 24 h postinfection (hpi). Knockdown of OASL increased the level of NDV viral RNA at 4, 8, and 24 hpi (P < 0.05) and eliminated the difference between NDV-infected and noninfected cells for expression of interferon response- and apoptosis-related genes (P > 0.05). The lack of differential expression suggests that knockdown of OASL resulted in a decreased response to NDV infection. Within NDV-infected cells, OASL knockdown reduced expression of signal transducer and activator of transcription 1, interferon alfa receptor subunit 1, eukaryotic translation initiation factor 2 alpha kinase 2, ribonuclease L, caspase 8 (CASP8) and caspase 9 (CASP9) at 4 hpi, CASP9 at 8 hpi, and caspase 3, CASP8, and CASP9 at 24 hpi (P < 0.05). We suggest that the increased NDV viral load in DF-1 cells after OASL knockdown was the result of a complex interaction between OASL and interferon response- and apoptosis-related genes that decreased host response to the NDV. Our results provide comprehensive information on the role played by OASL during NDV infection in vitro. Targeting this mechanism could aid in future prophylactic and therapeutic treatments for Newcastle disease in poultry.
Collapse
Affiliation(s)
- Ana Paula Del Vesco
- Department of Animal Science, Iowa State University, 50011-3150 Ames, USA; Department of Animal Science, Universidade Federal de Sergipe, 49100-000 São Cristóvão, Sergipe, Brazil
| | - Hyun Jun Jang
- Department of Animal Science, Iowa State University, 50011-3150 Ames, USA; Department of Animal Biotechnology, Jeonbuk National University, Jeonju-si, Jeollabuk-do 54896, Republic of Korea; Center for Industrialization of Agricultural and Livestock Microorganisms, Jeongeup-si, Jeollabuk-do 56212, Republic of Korea
| | - Melissa S Monson
- Department of Animal Science, Iowa State University, 50011-3150 Ames, USA
| | - Susan J Lamont
- Department of Animal Science, Iowa State University, 50011-3150 Ames, USA.
| |
Collapse
|
18
|
Ni J, Hu S, Wang X, Liu X, Hu Z, Liu X. Activation of the extracellular signal-regulated kinase pathway is required for replication of Newcastle disease virus. Arch Virol 2021; 166:921-927. [PMID: 33486628 DOI: 10.1007/s00705-020-04938-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/11/2020] [Indexed: 11/30/2022]
Abstract
Replication of Newcastle disease virus (NDV) is regulated by various host mechanisms, but the role of the extracellular signal-regulated kinase (ERK) pathway in regulating NDV replication is an open question. In this study, the relationship between the ERK pathway and NDV replication was investigated. NDV activated the ERK signaling in chicken embryo fibroblasts at the late stage of infection, correlating to expression of viral proteins. Specific blockage of the ERK pathway activation significantly decreased the transcription and translation levels of viral genes as well as virus replication and the cytopathogenic effect caused by NDV. Our results demonstrate that activation of the ERK pathway is required for NDV replication.
Collapse
Affiliation(s)
- Jie Ni
- Key Laboratory of Animal Infectious Diseases, School of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China
| | - Shunlin Hu
- Key Laboratory of Animal Infectious Diseases, School of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-Food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, No. 12, East Wenhui Rd, Yangzhou, 225009, Jiangsu, China
| | - Xiaoquan Wang
- Key Laboratory of Animal Infectious Diseases, School of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-Food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, No. 12, East Wenhui Rd, Yangzhou, 225009, Jiangsu, China
| | - Xiaowen Liu
- Key Laboratory of Animal Infectious Diseases, School of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-Food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, 225009, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, No. 12, East Wenhui Rd, Yangzhou, 225009, Jiangsu, China
| | - Zenglei Hu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China. .,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, No. 12, East Wenhui Rd, Yangzhou, 225009, Jiangsu, China.
| | - Xiufan Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China. .,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, No. 12, East Wenhui Rd, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
19
|
Jadhav A, Zhao L, Liu W, Ding C, Nair V, Ramos-Onsins SE, Ferretti L. Genomic Diversity and Evolution of Quasispecies in Newcastle Disease Virus Infections. Viruses 2020; 12:v12111305. [PMID: 33202558 PMCID: PMC7698180 DOI: 10.3390/v12111305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/31/2020] [Accepted: 11/06/2020] [Indexed: 12/19/2022] Open
Abstract
Newcastle disease virus (NDV) infections are well known to harbour quasispecies, due to the error-prone nature of the RNA polymerase. Quasispecies variants in the fusion cleavage site of the virus are known to significantly change its virulence. However, little is known about the genomic patterns of diversity and selection in NDV viral swarms. We analyse deep sequencing data from in vitro and in vivo NDV infections to uncover the genomic patterns of diversity and the signatures of selection within NDV swarms. Variants in viruses from in vitro samples are mostly localised in non-coding regions and 3′ and 5′ untranslated regions (3′UTRs or 5′UTRs), while in vivo samples contain an order of magnitude more variants. We find different patterns of genomic divergence and diversity among NDV genotypes, as well as differences in the genomic distribution of intra-host variants among in vitro and in vivo infections of the same strain. The frequency spectrum shows clear signatures of intra-host purifying selection in vivo on the matrix protein (M) coding gene and positive or diversifying selection on nucleocapsid (NP) and haemagglutinin-neuraminidase (HN). The comparison between within-host polymorphisms and phylogenetic divergence reveals complex patterns of selective pressure on the NDV genome at between- and within-host level. The M sequence is strongly constrained both between and within hosts, fusion protein (F) coding gene is under intra-host positive selection, and NP and HN show contrasting patterns: HN RNA sequence is positively selected between hosts while its protein sequence is positively selected within hosts, and NP is under intra-host positive selection at the RNA level and negative selection at the protein level.
Collapse
Affiliation(s)
- Archana Jadhav
- Viral Oncogenesis Group, The Pirbright Institute, Pirbright, Woking GU24 0NF, Surrey, UK; (A.J.); (V.N.)
| | - Lele Zhao
- Nuffield Department of Medicine, Li Ka Shing Centre for Health Information and Discovery, Big Data Institute, University of Oxford, Oxford OX3 7LF, UK;
| | - Weiwei Liu
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (W.L.); (C.D.)
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (W.L.); (C.D.)
| | - Venugopal Nair
- Viral Oncogenesis Group, The Pirbright Institute, Pirbright, Woking GU24 0NF, Surrey, UK; (A.J.); (V.N.)
- UK-China Centre of Excellence on Avian Disease Research, Pirbright, Woking GU24 0NF, Surrey, UK
| | - Sebastian E. Ramos-Onsins
- Plant and Animal Genomics, Centre de Recerca en Agrigenòmica (CRAG) CSIC-IRTA-UAB-UB, 08193 Bellaterra, Spain
- Correspondence: (S.E.R.-O.); (L.F.)
| | - Luca Ferretti
- Nuffield Department of Medicine, Li Ka Shing Centre for Health Information and Discovery, Big Data Institute, University of Oxford, Oxford OX3 7LF, UK;
- Correspondence: (S.E.R.-O.); (L.F.)
| |
Collapse
|
20
|
Rao PL, Gandham RK, Subbiah M. Molecular evolution and genetic variations of V and W proteins derived by RNA editing in Avian Paramyxoviruses. Sci Rep 2020; 10:9532. [PMID: 32533018 PMCID: PMC7293227 DOI: 10.1038/s41598-020-66252-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 05/06/2020] [Indexed: 11/12/2022] Open
Abstract
The newly assigned subfamily Avulavirinae in the family Paramyxoviridae includes avian paramyxoviruses (APMVs) isolated from a wide variety of avian species across the globe. Till date, 21 species of APMVs are reported and their complete genome sequences are available in GenBank. The APMV genome comprises of a single stranded, negative sense, non-segmented RNA comprising six transcriptional units (except APMV-6 with seven units) each coding for a structural protein. Additionally, by co-transcriptional RNA editing of phosphoprotein (P) gene, two mRNAs coding for accessory viral proteins, V and W, are generated along with unedited P mRNA. However, in APMV-11, the unedited mRNA codes for V protein while +2 edited mRNA translates to P protein, similar to members of subfamily Rubulavirinae in the same family. Such RNA editing in paramyxoviruses enables maximizing the coding capacity of their smaller genome. The three proteins of P gene: P, V and W, share identical N terminal but varied C terminal sequences that contribute to their unique functions. Here, we analyzed the P gene editing site, V and W sequences of all 21 APMV species known so far (55 viruses) by using bioinformatics and report their genetic variations and molecular evolution. The variations observed in the sequence and hexamer phase positions of the P gene editing sites is likely to influence the levels and relative proportions of P, V and W proteins' expressions which could explain the differences in the pathogenicity of APMVs. The V protein sequences of APMVs had conserved motifs similar to V proteins of other paramyxoviruses including the seven cysteine residues involved in MDA5 interference, STAT1 degradation and interferon antagonism. Conversely, W protein sequences of APMVs were distinct. High sequence homology was observed in both V and W proteins between strains of the same species than between species except in APMV-3 which was the most divergent APMV species. The estimates of synonymous and non-synonymous substitution rates suggested negative selection pressure on the V and W proteins within species indicating their low evolution rate. The molecular clock analysis revealed higher conservation of V protein sequence compared to W protein indicating the important role played by V protein in viral replication, pathogenesis and immune evasion. However, we speculate the genetic diversity of W proteins could impact the degree of pathogenesis, variable interferon antagonistic activity and the wide host range exhibited by APMV species. Phylogenetically, V proteins of APMVs clustered into three groups similar to the recent classification of APMVs into three new genera while no such pattern could be deciphered in the analysis of W proteins except that strains of same species grouped together. This is the first comprehensive study describing in detail the genetic variations and the molecular evolution of P gene edited, accessory viral proteins of Avian paramyxoviruses.
Collapse
Affiliation(s)
| | - Ravi Kumar Gandham
- National Institute of Animal Biotechnology, Hyderabad, 500032, Telangana, India
| | - Madhuri Subbiah
- National Institute of Animal Biotechnology, Hyderabad, 500032, Telangana, India.
| |
Collapse
|
21
|
Yan C, Liu H, Jia Y, Prince-Theodore DW, Yang M, Addoma Adam FE, Ren J, Cao X, Wang X, Xiao S, Zhang S, Yang Z. Screening and mechanistic study of key sites of the hemagglutinin-neuraminidase protein related to the virulence of Newcastle disease virus. Poult Sci 2020; 99:3374-3384. [PMID: 32616231 PMCID: PMC7597932 DOI: 10.1016/j.psj.2020.04.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/03/2020] [Accepted: 04/08/2020] [Indexed: 11/25/2022] Open
Abstract
Newcastle disease is a kind of avian infectious disease caused by Newcastle disease virus (NDV). The virulence of NDV is dependent mainly on the fusion (F) protein and hemagglutinin-neuraminidase (HN) protein. The genomes of 2 viruses, NDV-Blackbird and NDV-Dove, are 99.9% similar, while NDV-Blackbird is a velogenic virus, and NDV-Dove is a lentogenic virus. Further analysis revealed that the F proteins of the 2 strains were identical, and only 5 amino acid sites on the HN proteins were inconsistent. Five different HN mutant plasmids were constructed and analyzed in this study. The results showed that the mutation F110L caused a significant increase in fusion-promotion activity caused by an increase in neuraminidase activity. Because of the increase in receptor-binding activity caused by G116R, there was a significant increase in fusion-promotion activity. The mutation G54S resulted in a slight decrease in the fusion-promotion activity caused by a slight decrease in receptor-binding activity. The slight increase in the fusion-promotion activity caused by A469V was associated with a significant increase in neuraminidase activity. Therefore, the amino acids L110 and R116 played a key role in determining the virulence difference between NDV-Blackbird and NDV-Dove, which could lay a foundation for illuminating the virulence differences of NDV strains, as well as the development of attenuated vaccines.
Collapse
Affiliation(s)
- Chuanqi Yan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, China
| | - Haijin Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, China
| | - Yanqing Jia
- Department of Animal Engineering, Yangling Vocational & Technical College, Yangling 712100, Shaanxi Province, China
| | - Daguia-Wenam Prince-Theodore
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, China; Institut Superieur du Developpement Rural (ISDR), Universite de Bangui BP 1450, Bangui, Republique Centrafricaine
| | - Mengqing Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, China
| | - Fathalrhman Eisa Addoma Adam
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, China; Department of Preventive Medicine and Public Health, Faculty of Veterinary Science, University of Nyala, Nyala, Sudan
| | - Juan Ren
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, China
| | - Xuhong Cao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, China
| | - Xinglong Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, China
| | - Sa Xiao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, China
| | - Shuxia Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, China.
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi Province, China.
| |
Collapse
|
22
|
Yang Y, Bu Y, Zhao J, Xue J, Xu G, Song Y, Zhao Y, Yang H, Zhang G. Appropriate amount of W protein of avian avulavirus 1 benefits viral replication and W shows strain-dependent subcellular localization. Virology 2019; 538:71-85. [PMID: 31580973 DOI: 10.1016/j.virol.2019.09.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/17/2019] [Accepted: 09/26/2019] [Indexed: 11/28/2022]
Abstract
In order to confirm the existence of W protein in Avian avulavirus 1 (AAvV-1) infected cells, two monoclonal antibodies were prepared. The presence of W protein in cells infected with lentogenic genotype II strain La Sota or velogenic genotype VII strain SG10 was confirmed with immunofluorescence and western blotting assays. WSG10 localized to the cytoplasm, whereas WLa Sota localized to the nucleus. The influence of W protein was investigated in vitro and in vivo with two AAvV-1 strains defective in the W C-terminus. The growth kinetic curves and pathogenicity tests in 3-week-old SPF chickens both showed that the replication abilities of strains with C-terminally deleted W proteins were lower than that of the parental strain. Restoring the appropriate dose of W protein increased the viral titers of these strains. The expression validation and functional exploration of W protein will facilitate our understanding of pathogenic mechanism of AAvV-1.
Collapse
Affiliation(s)
- Yanling Yang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Yawen Bu
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jing Zhao
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jia Xue
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Gang Xu
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Yang Song
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Ye Zhao
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Huiming Yang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Guozhong Zhang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
23
|
Chu Z, Gao X, Liu H, Ma J, Wang C, Lu K, Han Q, Wang Y, Wang C, Adam FEA, Wang X, Xiao S, Yang Z. Newcastle disease virus selectively infects dividing cells and promotes viral proliferation. Vet Res 2019; 50:27. [PMID: 30999941 PMCID: PMC6472075 DOI: 10.1186/s13567-019-0644-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 01/23/2019] [Indexed: 12/15/2022] Open
Abstract
Newcastle disease virus (NDV) can select cells to infect, but the mechanism of its cell selectivity has not been comprehensively investigated. Here, we use HeLa cells to establish that NDV can selectively infect cells at the single-cell level. We labeled proliferating cells with 5′-bromo-2-deoxyuridine (BrdU) and examined the colocalization of BrdU with NDV in cells to clarify the relationships between NDV infection and cell proliferation. Receptors at the plasma membrane mediate NDV entry into host cells. We labeled sialic acid receptor isoforms, compared their densities between different cell types and measured the sialic acid receptor densities in different cell phases. Our results suggest that NDV displays host tropism to HeLa cells compared to BHK cells and that the differences in the receptor isoform expression patterns between cell types contribute to the selection of HeLa by NDV. At the single-cell level, the dynamics of receptor expression changes during different cell phases contributing to the selection of cells in S/G2 phase for NDV infection. Furthermore, cell proliferation benefits viral replication, and enhanced virus replication leads to increased damage to cells. The elucidation of the mechanisms underlying host cell selection by NDV may help in the screening and characterizing of additional candidate oncolytic virus strains.
Collapse
Affiliation(s)
- Zhili Chu
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100, Shaanxi, People's Republic of China.,School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People's Republic of China
| | - Xiaolong Gao
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Haijin Liu
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Jiangang Ma
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Caiying Wang
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Kejia Lu
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Qingsong Han
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Yanhong Wang
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Chongyang Wang
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Fathalrhman E A Adam
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100, Shaanxi, People's Republic of China.,Department of Preventive Medicine and Public Health, Faculty of Veterinary Science, University of Nyala, P.O Box: 155, Nyala, Sudan
| | - Xinglong Wang
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Sa Xiao
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100, Shaanxi, People's Republic of China
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100, Shaanxi, People's Republic of China.
| |
Collapse
|
24
|
Wang C, Chu Z, Liu W, Pang Y, Gao X, Tang Q, Ma J, Lu K, Adam FEA, Dang R, Xiao S, Wang X, Yang Z. Newcastle disease virus V protein inhibits apoptosis in DF-1 cells by downregulating TXNL1. Vet Res 2018; 49:102. [PMID: 30290847 PMCID: PMC6389150 DOI: 10.1186/s13567-018-0599-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 09/08/2018] [Indexed: 02/06/2023] Open
Abstract
Many viral proteins are related to suppressing apoptosis in target cells and are hence beneficial to viral replication. The V protein of Newcastle disease virus (NDV) is one such protein that plays an important role in inhibiting apoptosis in a species-specific manner. However, to date, there have been no reports clarifying the antiapoptotic mechanisms of the V protein. The present study was undertaken to determine the apoptotic potential of the V protein in a chicken embryo fibroblast cell line (DF-1 cell) and to elucidate its molecular mechanisms of action. Here, a yeast two-hybrid system was used to screen the host proteins that interact with the V protein and identified thioredoxin-like protein 1 (TXNL1) as a potential binding partner. Immuno-colocalization of V protein and TXNL1 protein in DF-1 cells further verified the interaction of the two proteins. Through the overexpression of TXNL1 protein and knockdown of TXNL1 protein in DF-1 cells, the effects of NDV replication and cell apoptosis were examined. Cell apoptosis was detected by flow cytometry. The mRNA and protein expression levels of Bax, Bcl-2 and Caspase-3 were detected by quantitative real-time PCR (Q-PCR) and Western blotting. NDV expression was detected by Q-PCR and plaque assay. The results revealed that the TXNL1 protein induced apoptosis and inhibited NDV replication in DF-1 cells. Furthermore, the Western blot and Q-PCR results suggested that TXNL1 induced cell apoptosis through a pathway involving Bcl-2\Bax and Caspase-3. Finally, this work provides insight into the mechanism by which the V protein inhibits apoptosis.
Collapse
Affiliation(s)
- Caiying Wang
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100 Shaanxi China
| | - Zhili Chu
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100 Shaanxi China
| | - Wenkai Liu
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100 Shaanxi China
| | - Yu Pang
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100 Shaanxi China
| | - Xiaolong Gao
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100 Shaanxi China
| | - Qiuxia Tang
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100 Shaanxi China
| | - Jiangang Ma
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100 Shaanxi China
| | - Kejia Lu
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100 Shaanxi China
| | - Fathalrhman E. A. Adam
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100 Shaanxi China
- Department of Preventive Medicine and Public Health, Faculty of Veterinary Science, University of Nyala, P.O Box: 155, Nyala, Sudan
| | - Ruyi Dang
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100 Shaanxi China
| | - Sa Xiao
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100 Shaanxi China
| | - Xinglong Wang
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100 Shaanxi China
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A & F University, Yangling, 712100 Shaanxi China
| |
Collapse
|
25
|
Newcastle Disease Virus V Protein Promotes Viral Replication in HeLa Cells through the Activation of MEK/ERK Signaling. Viruses 2018; 10:v10090489. [PMID: 30213106 PMCID: PMC6163439 DOI: 10.3390/v10090489] [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: 08/02/2018] [Revised: 08/25/2018] [Accepted: 09/05/2018] [Indexed: 12/12/2022] Open
Abstract
Newcastle disease virus (NDV) can infect a wide range of domestic and wild bird species. The non-structural V protein of NDV plays an important role in antagonizing innate host defenses to facilitate viral replication. However, there is a lack of knowledge related to the mechanisms through which the V protein regulates viral replication. The extracellular signal-regulated kinase (ERK) signaling pathway in the host is involved in a variety of functions and is activated by several stimuli, including viral replication. In this study, we show that both the lentogenic strain, La Sota, and the velogenic strain, F48E9, of NDV activate the mitogen-activated protein kinase (MEK)/ERK signaling pathway. The pharmacological inhibition of ERK1/2 phosphorylation using the highly selective inhibitors U0126 and SCH772984 resulted in the reduced levels of NDV RNA in cells and virus titers in the cell supernatant, which established an important role for the MEK/ERK signaling pathway in NDV replication. Moreover, the overexpression of the V protein in HeLa cells increased the phosphorylation of ERK1/2 and induced the transcriptional changes in the genes downstream of the MEK/ERK signaling pathway. Taken together, our results demonstrate that the V protein is involved in the ERK signaling pathway-mediated promotion of NDV replication and thus, can be investigated as a potential antiviral target.
Collapse
|