1
|
Mishra S, Pandey A, Verma J, Rajala MS. Analysis of the interaction of influenza a virus nucleoprotein with host cell nucleolin. Arch Virol 2024; 170:1. [PMID: 39636515 DOI: 10.1007/s00705-024-06189-y] [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: 03/13/2024] [Accepted: 09/25/2024] [Indexed: 12/07/2024]
Abstract
Targeting interactions between a virus and a host protein is one of the important approaches to developing antiviral therapies. We previously identified host nucleolin as a novel interacting partner of the influenza A virus nucleoprotein, and it was demonstrated that this interaction restricts virus replication. In the current study, we examined the interaction of nucleolin with the viral nucleoprotein at the domain and amino acid levels using in vitro and in silico approaches. Both approaches demonstrated a direct and specific interaction between these two proteins. Furthermore, it was observed that previous pandemic strains of influenza A virus had specific amino acid residues in their nucleoproteins that were predicted to be critical for interaction with nucleolin. This preliminary analysis provides insights into the binding process, which could be explored for developing antiviral strategies.
Collapse
Affiliation(s)
- Shruti Mishra
- School of Biotechnology, Jawaharlal Nehru University, 110067, New Delhi, India
- School of Biological and Life sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Achyut Pandey
- School of Biotechnology, Jawaharlal Nehru University, 110067, New Delhi, India
- School of Biomedical Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Jyoti Verma
- School of Computational and integrative Sciences, Jawaharlal Nehru University, 110067, New Delhi, India
| | - Maitreyi S Rajala
- School of Biotechnology, Jawaharlal Nehru University, 110067, New Delhi, India.
| |
Collapse
|
2
|
Panthi S, Hong JY, Satange R, Yu CC, Li LY, Hou MH. Antiviral drug development by targeting RNA binding site, oligomerization and nuclear export of influenza nucleoprotein. Int J Biol Macromol 2024; 282:136996. [PMID: 39486729 DOI: 10.1016/j.ijbiomac.2024.136996] [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: 08/12/2024] [Revised: 10/23/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
The quasispecies of the influenza virus poses a significant challenge for developing effective therapies. Current antiviral drugs such as oseltamivir, zanamivir, peramivir and baloxavir marboxil along with seasonal vaccines have limitations due to viral variability caused by antigenic drift and shift as well as the development of drug resistance. Therefore, there is a clear need for novel antiviral agents targeting alternative mechanisms, either independently or in combination with existing modalities, to reduce the impact of influenza virus-related infections. The influenza nucleoprotein (NP) is a key component of the viral ribonucleoprotein complex. The multifaceted nature of the NP makes it an attractive target for antiviral intervention. Recent reports have identified inhibitors that specifically target this protein. Recognizing the importance of developing influenza treatments for potential pandemics, this review explores the structural and functional aspects of NP and highlights its potential as an emerging target for anti-influenza drugs. We discuss various strategies for targeting NP, including RNA binding, oligomerization, and nuclear export, and also consider the potential of NP-based vaccines. Overall, this review provides insights into recent developments and future perspectives on targeting influenza NP for antiviral therapies.
Collapse
Affiliation(s)
- Sankar Panthi
- Doctoral Program in Medical Biotechnology, National Chung Hsing University, Taichung 402, Taiwan; Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
| | - Jhen-Yi Hong
- Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Roshan Satange
- Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
| | - Ching-Ching Yu
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Long-Yuan Li
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan.
| | - Ming-Hon Hou
- Doctoral Program in Medical Biotechnology, National Chung Hsing University, Taichung 402, Taiwan; Graduate Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan; Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan; Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan.
| |
Collapse
|
3
|
Gu M, Jiao J, Liu S, Zhao W, Ge Z, Cai K, Xu L, He D, Zhang X, Qi X, Jiang W, Zhang P, Wang X, Hu S, Liu X. Monoclonal antibody targeting a novel linear epitope on nucleoprotein confers pan-reactivity to influenza A virus. Appl Microbiol Biotechnol 2023; 107:2437-2450. [PMID: 36820898 PMCID: PMC9947902 DOI: 10.1007/s00253-023-12433-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/20/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023]
Abstract
Nucleoprotein (NP) functions crucially in the replicative cycle of influenza A virus (IAV) via forming the ribonucleoprotein complex together with PB2, PB1, and PA proteins. As its high conservation, NP ranks one of the hot targets for design of universal diagnostic reagents and antiviral drugs for IAV. Here, we report an anti-NP murine monoclonal antibody (mAb) 5F10 prepared from traditional lymphocyte hybridoma technique with the immunogen of a clade 2.3.4.4 H5N1 subtype avian influenza virus. The specificity of mAb 5F10 to NP protein was confirmed by immunofluorescence assay and western blotting, and the mAb 5F10 could be used in immunoprecipitation and immunohistochemistry assays. Importantly, mAb 5F10 possessed broad-spectrum reactivity against H1~H11 subtypes of avian influenza viruses, including various HA clades of H5Nx subtype. In addition, mAb 5F10 also showed good affinity with H1N1 and H3N2 subtype influenza viruses of swine and human origin. Furthermore, the recognized antigenic epitope of mAb 5F10 was identified to consist of the conserved amino acid motif 81EHPSA85 in the second flexible loop region of NP protein through screening the phage display peptide library. Collectively, the mAb 5F10 which recognizes the novel universal NP linear B-cell epitope of IAV with diverse origins and subtypes will be a powerful tool for NP protein-based structural, functional, and mechanistic studies, as well as the development of detection methods and universal vaccines for IAV. KEY POINTS: • A broad-spectrum mAb against various subtypes and sources of IAV was developed • The mAb possessed good reactivity in IFA, western blot, IP, and IHC assays • The mAb targeted a novel conserved linear B-cell epitope involving 81EHPSA85 on NP protein.
Collapse
Affiliation(s)
- Min Gu
- grid.268415.cAnimal Infectious Diseases Laboratory, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009 Jiangsu China
- grid.268415.cJiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu China
- grid.268415.cJiangsu Key Laboratory of Zoonoses, Yangzhou University, Yangzhou, 225009 Jiangsu China
| | - Jun Jiao
- grid.268415.cAnimal Infectious Diseases Laboratory, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009 Jiangsu China
| | - Suhan Liu
- grid.268415.cAnimal Infectious Diseases Laboratory, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009 Jiangsu China
| | - Wanchen Zhao
- grid.268415.cAnimal Infectious Diseases Laboratory, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009 Jiangsu China
| | - Zhichuang Ge
- grid.268415.cAnimal Infectious Diseases Laboratory, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009 Jiangsu China
| | - Kairui Cai
- grid.268415.cAnimal Infectious Diseases Laboratory, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009 Jiangsu China
| | - Lijun Xu
- grid.268415.cAnimal Infectious Diseases Laboratory, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009 Jiangsu China
| | - Dongchang He
- grid.268415.cAnimal Infectious Diseases Laboratory, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009 Jiangsu China
| | - Xinyu Zhang
- grid.268415.cAnimal Infectious Diseases Laboratory, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009 Jiangsu China
| | - Xian Qi
- grid.410734.50000 0004 1761 5845Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009 China
| | - Wenming Jiang
- grid.414245.20000 0004 6063 681XChina Animal Health and Epidemiology Center, Qingdao, 266032 China
| | - Pinghu Zhang
- grid.268415.cJiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu China
- grid.268415.cJiangsu Key Laboratory of Zoonoses, Yangzhou University, Yangzhou, 225009 Jiangsu China
| | - Xiaoquan Wang
- grid.268415.cAnimal Infectious Diseases Laboratory, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009 Jiangsu China
- grid.268415.cJiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu China
- grid.268415.cJiangsu Key Laboratory of Zoonoses, Yangzhou University, Yangzhou, 225009 Jiangsu China
| | - Shunlin Hu
- grid.268415.cAnimal Infectious Diseases Laboratory, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009 Jiangsu China
- grid.268415.cJiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu China
- grid.268415.cJiangsu Key Laboratory of Zoonoses, Yangzhou University, Yangzhou, 225009 Jiangsu China
| | - Xiufan Liu
- grid.268415.cAnimal Infectious Diseases Laboratory, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009 Jiangsu China
- grid.268415.cJiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 Jiangsu China
- grid.268415.cJiangsu Key Laboratory of Zoonoses, Yangzhou University, Yangzhou, 225009 Jiangsu China
| |
Collapse
|
4
|
Cui H, Zhang C, Tu F, Zhao K, Kong Y, Pu J, Zhang L, Chen Z, Sun Y, Wei Y, Liang C, Liu J, Liu J, Guo Z. Rapid detection of influenza A viruses using a real-time reverse transcription recombinase-aided amplification assay. Front Cell Infect Microbiol 2023; 12:1071288. [PMID: 36683681 PMCID: PMC9849684 DOI: 10.3389/fcimb.2022.1071288] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction Influenza A viruses (IAVs) are important pathogens of respiratory infections, causing not only seasonal influenza but also influenza pandemics and posing a global threat to public health. IAVs infection spreads rapidly, widely, and across species, causing huge losses, especially zoonotic IAVs infections that are more harmful. Fast and sensitive detection of IAVs is critical for controlling the spread of this disease. Methods Here, a real-time reverse transcription recombinase-aided amplification (real-time RT-RAA) assay targeting conserved positions in the matrix protein gene (M gene) of IAVs, is successfully established to detect IAVs. The assay can be completed within 20 min at 42°C. Results The sensitivity of the real-time RT-RAA assay was 142 copies per reaction at 95% probability, which was comparable to the sensitivity of the RT-qPCR assay. The specificity assay showed that the real-time RT-RAA assay was specific to IAVs, and there was no cross-reactivity with other important viruses. In addition, 100%concordance between the real-time RT-RAA and RT-qPCR assays was achieved after testing 120 clinical specimens. Discussion The results suggested that the real-time RT-RAA assay we developed was a specific, sensitive and reliable diagnostic tool for the rapid detection of IAVs.
Collapse
Affiliation(s)
- Huan Cui
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
- College of Animal Medicine, Jilin University, Changchun, China
| | - Cheng Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Fei Tu
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
| | - Kui Zhao
- College of Animal Medicine, Jilin University, Changchun, China
| | - Yunyi Kong
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
| | - Jie Pu
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
| | - Lei Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
| | - Zhaoliang Chen
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Yuanyuan Sun
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Yujie Wei
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Chuncai Liang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Juxiang Liu
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Jun Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
| | - Zhendong Guo
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
| |
Collapse
|
5
|
An C, Wu Y, Wu J, Liu H, Zhou S, Ge D, Dong R, You L, Hao Y. Berberine ameliorates pulmonary inflammation in mice with influenza viral pneumonia by inhibiting NLRP3 inflammasome activation and gasdermin D‐mediated pyroptosis. Drug Dev Res 2022; 83:1707-1721. [DOI: 10.1002/ddr.21995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/10/2022] [Accepted: 08/23/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Chen An
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Yanmin Wu
- Department of Immunology, School of Medical Technology Qiqihar Medical University Qiqihar China
| | - Jun Wu
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Huanwei Liu
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Siyao Zhou
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Dongyu Ge
- Research and Test Center, School of Traditional Chinese Medicine Beijing University of Chinese Medicine Beijing China
| | - Ruijuan Dong
- Research and Test Center, School of Traditional Chinese Medicine Beijing University of Chinese Medicine Beijing China
| | - Leiming You
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Yu Hao
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| |
Collapse
|
6
|
Mostafa A, Blaurock C, Scheibner D, Müller C, Blohm U, Schäfer A, Gischke M, Salaheldin AH, Nooh HZ, Ali MA, Breithaupt A, Mettenleiter TC, Pleschka S, Abdelwhab EM. Genetic incompatibilities and reduced transmission in chickens may limit the evolution of reassortants between H9N2 and panzootic H5N8 clade 2.3.4.4 avian influenza virus showing high virulence for mammals. Virus Evol 2020; 6:veaa077. [PMID: 33343923 PMCID: PMC7733613 DOI: 10.1093/ve/veaa077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The unprecedented spread of H5N8- and H9N2-subtype avian influenza virus (AIV) in birds across Asia, Europe, Africa, and North America poses a serious public health threat with a permanent risk of reassortment and the possible emergence of novel virus variants with high virulence in mammals. To gain information on this risk, we studied the potential for reassortment between two contemporary H9N2 and H5N8 viruses. While the replacement of the PB2, PA, and NS genes of highly pathogenic H5N8 by homologous segments from H9N2 produced infectious H5N8 progeny, PB1 and NP of H9N2 were not able to replace the respective segments from H5N8 due to residues outside the packaging region. Furthermore, exchange of the PB2, PA, and NS segments of H5N8 by those of H9N2 increased replication, polymerase activity and interferon antagonism of the H5N8 reassortants in human cells. Notably, H5N8 reassortants carrying the H9N2-subtype PB2 segment and to lesser extent the PA or NS segments showed remarkably increased virulence in mice as indicated by rapid onset of mortality, reduced mean time to death and increased body weight loss. Simultaneously, we observed that in chickens the H5N8 reassortants, particularly with the H9N2 NS segment, demonstrated significantly reduced transmission to co-housed chickens. Together, while the limited capacity for reassortment between co-circulating H9N2 and H5N8 viruses and the reduced bird-to-bird transmission of possible H5N8 reassortants in chickens may limit the evolution of such reassortant viruses, they show a higher replication potential in human cells and increased virulence in mammals.
Collapse
Affiliation(s)
| | | | | | - Christin Müller
- Institute of Medical Virology, Justus Liebig University Giessen, Schubertstrasse 81, 35392 Giessen, Germany
| | - Ulrike Blohm
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany
| | - Alexander Schäfer
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany
| | | | | | - Hanaa Z Nooh
- Department of Anatomy and Histology, College of Medicine, Jouf University, Sakaka 72442, Aljouf Province, Saudi Arabia
| | - Mohamed A Ali
- Center of Scientific Excellence for Influenza Viruses, National Research Centre (NRC), Dokki, 12622, Giza, Egypt
| | - Angele Breithaupt
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany
| | | | - Stephan Pleschka
- Institute of Medical Virology, Justus Liebig University Giessen, Schubertstrasse 81, 35392 Giessen, Germany
| | | |
Collapse
|
7
|
Dissecting the mechanism of signaling-triggered nuclear export of newly synthesized influenza virus ribonucleoprotein complexes. Proc Natl Acad Sci U S A 2020; 117:16557-16566. [PMID: 32601201 PMCID: PMC7368312 DOI: 10.1073/pnas.2002828117] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Influenza viruses (IV) replicate in the nucleus. Export of newly produced genomes, packaged in viral ribonucleoprotein (vRNP) complexes, relies on the nuclear CRM1 export pathway and appears to be timely controlled by virus-induced cellular signaling. However, the exact mechanism of the signaling-controlled complex assembly and export is enigmatic. Here we show that IV activates the Raf/MEK/ERK/RSK1 pathway, leading to phosphorylation at specific sites of the NP, which in turn, creates a docking site for binding of the M1 protein, an initial step in formation of vRNP export complexes. These findings are of broad relevance regarding the regulatory role of signaling pathways and posttranslational modifications in virus propagation and will strongly support ongoing development of an alternative anti-influenza therapy. Influenza viruses (IV) exploit a variety of signaling pathways. Previous studies showed that the rapidly accelerated fibrosarcoma/mitogen-activated protein kinase/extracellular signal-regulated kinase (Raf/MEK/ERK) pathway is functionally linked to nuclear export of viral ribonucleoprotein (vRNP) complexes, suggesting that vRNP export is a signaling-induced event. However, the underlying mechanism remained completely enigmatic. Here we have dissected the unknown molecular steps of signaling-driven vRNP export. We identified kinases RSK1/2 as downstream targets of virus-activated ERK signaling. While RSK2 displays an antiviral role, we demonstrate a virus-supportive function of RSK1, migrating to the nucleus to phosphorylate nucleoprotein (NP), the major constituent of vRNPs. This drives association with viral matrix protein 1 (M1) at the chromatin, important for vRNP export. Inhibition or knockdown of MEK, ERK or RSK1 caused impaired vRNP export and reduced progeny virus titers. This work not only expedites the development of anti-influenza strategies, but in addition demonstrates converse actions of different RSK isoforms.
Collapse
|
8
|
Jin Z, Wang Y, Yu XF, Tan QQ, Liang SS, Li T, Zhang H, Shaw PC, Wang J, Hu C. Structure-based virtual screening of influenza virus RNA polymerase inhibitors from natural compounds: Molecular dynamics simulation and MM-GBSA calculation. Comput Biol Chem 2020; 85:107241. [DOI: 10.1016/j.compbiolchem.2020.107241] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 02/22/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022]
|
9
|
Morris AK, Wang Z, Ivey AL, Xie Y, Hill PS, Schey KL, Ren Y. Cellular mRNA export factor UAP56 recognizes nucleic acid binding site of influenza virus NP protein. Biochem Biophys Res Commun 2020; 525:259-264. [PMID: 32085897 DOI: 10.1016/j.bbrc.2020.02.059] [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: 01/03/2020] [Accepted: 02/09/2020] [Indexed: 02/06/2023]
Abstract
Influenza A virus nucleoprotein (NP) is a structural component that encapsulates the viral genome into the form of ribonucleoprotein complexes (vRNPs). Efficient assembly of vRNPs is critical for the virus life cycle. The assembly route from RNA-free NP to the NP-RNA polymer in vRNPs has been suggested to require a cellular factor UAP56, but the mechanism is poorly understood. Here, we characterized the interaction between NP and UAP56 using recombinant proteins and showed that UAP56 features two NP binding sites. In addition to the UAP56 core comprised of two RecA domains, we identified the N-terminal extension (NTE) of UAP56 as a previously unknown NP binding site. In particular, UAP56-NTE recognizes the nucleic acid binding region of NP. This corroborates our observation that binding of UAP56-NTE and RNA to NP is mutually exclusive. Collectively, our results reveal the molecular basis for how UAP56 acts on RNA-free NP, and provide new insights into NP-mediated influenza genome packaging.
Collapse
Affiliation(s)
- Andrew K Morris
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Zhen Wang
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Austin L Ivey
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Yihu Xie
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Pate S Hill
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Kevin L Schey
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Yi Ren
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
| |
Collapse
|
10
|
Toro-Ascuy D, Santibañez A, Peña V, Beltran-Pavez C, Cottet L, Molina C, Montoya M, Sandoval N, Vásquez-Martínez Y, Mascayano C, Cortez-San Martín M. Development of an Isavirus minigenome system to study the function of the pocket RNA-binding domain of the viral nucleoprotein (NP) in salmon cells. JOURNAL OF FISH DISEASES 2020; 43:197-206. [PMID: 31845350 DOI: 10.1111/jfd.13122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 06/10/2023]
Abstract
The Isavirus is an orthomyxovirus with a genome composed of eight segments of negative single-strand RNA (-ssRNA). It has been proposed that the eight genomic segments of the Isavirus are organized as a ribonucleoprotein (RNP) complex called a minigenome, which contains all the viral RNA segments, a viral heterotrimeric polymerase and multiple copies of the viral nucleoprotein (NP). Here, we develop an Isavirus minigenome system and show the importance of the formation of active RNPs and the role of viral NP R189, R194, R302 and K325 residues in the NP RNA-binding domain in the context of RNPs. The results indicate it is possible to generate a minigenome in salmon cells, a composite ISAV RNPs with EGFP-based chimeric vRNA with heterotrimeric polymerase (PB1, PB2, PA) and NP protein using CMV-based auxiliary plasmids. It was also shown that NP R189, R194, R302 and K325 residues are important to generate viral mRNA from the constituted RNPs and a detectable reporter protein. This work is the first salmon cell-based minigenome assay for the Isavirus, which was evaluated by a bioinformatic and functional study of the NP protein in viral RNPs, which showed that correct NP-vRNA interaction is key to the functioning of RNPs.
Collapse
Affiliation(s)
- Daniela Toro-Ascuy
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomedicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Alvaro Santibañez
- Laboratory of Molecular Virology and Pathogen Control, Department of Biology, Faculty of Chemistry and Biology, University of Santiago, Santiago, Chile
| | - Victor Peña
- Laboratory of Molecular Virology and Pathogen Control, Department of Biology, Faculty of Chemistry and Biology, University of Santiago, Santiago, Chile
| | - Carolina Beltran-Pavez
- Laboratory of Molecular Virology and Pathogen Control, Department of Biology, Faculty of Chemistry and Biology, University of Santiago, Santiago, Chile
| | - Luis Cottet
- Laboratory of Molecular Virology and Pathogen Control, Department of Biology, Faculty of Chemistry and Biology, University of Santiago, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, University Santo Tomas, Santiago, Chile
| | - Cristian Molina
- Laboratory of Molecular Virology and Pathogen Control, Department of Biology, Faculty of Chemistry and Biology, University of Santiago, Santiago, Chile
| | - Margarita Montoya
- Cellular Biochemistry Laboratory, Department of Biology, Faculty of Chemistry and Biology, University of Santiago, Santiago, Chile
| | - Nicolas Sandoval
- Laboratory of Molecular Virology and Pathogen Control, Department of Biology, Faculty of Chemistry and Biology, University of Santiago, Santiago, Chile
| | - Yesseny Vásquez-Martínez
- Programa Centro de Investigaciones Biomédicas Aplicadas, Facultad de Ciencias Médicas, Universidad de Santiago, Santiago, Chile
| | - Carolina Mascayano
- Department of Environmental Sciences, Faculty of Chemistry and Biology, University of Santiago, Santiago, Chile
| | - Marcelo Cortez-San Martín
- Laboratory of Molecular Virology and Pathogen Control, Department of Biology, Faculty of Chemistry and Biology, University of Santiago, Santiago, Chile
| |
Collapse
|
11
|
Plotnikova MA, Klotchenko SA, Lebedev KI, Lozhkov AA, Taraskin AS, Gyulikhandanova NE, Ramsay ES, Vasin AV. Antibody microarray immunoassay for screening and differential diagnosis of upper respiratory tract viral pathogens. J Immunol Methods 2019; 478:112712. [PMID: 31783022 DOI: 10.1016/j.jim.2019.112712] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/12/2019] [Accepted: 11/25/2019] [Indexed: 01/26/2023]
Abstract
Upper respiratory tract infections are the world's most common infectious disease. The etiologic agents behind upper respiratory tract infections (URTIs) are, in fact, a diverse set of pathogens such as influenza, parainfluenza, adenovirus, rhinovirus, and others. More than 200 pathogens are known to be involved. Differential diagnosis of viral infections is sometimes complicated by their diversity or similarity of clinical presentation. This work is devoted to the development of a method which enables simultaneous detection of six common viral URTI pathogens: IAV; IBV; RSV; hAdV; hPIV2; and hPIV3. Antibody microarray technology is utilized to accomplish the analysis. In preparation for protein microchip creation, we produced, characterized, and selected approximately 50 monoclonal antibodies; for each of the aforementioned pathogens, an optimal monoclonal antibody pair was selected. A protein microchip was created, and its core working conditions were optimized. With a balance between convenience and maximal assay sensitivity in mind, a one-step analysis approach was developed for accomplishing the ELISA-like "sandwich" interaction on the manufactured microchip (antibody microarray). Reference viral strains were used to establish the lower limits of detection (LoD) for the assay. For IAV, the LoD was 0.25 ng/ml total viral protein. For other viruses, the LoD ranged from 1 to 2 ng/ml total protein. These sensitivity limits are slightly better than those of standard ELISA, but inferior to those of PCR. Overall, we believe that the developed microchip is a good alternative to existing methods, allowing relatively quick (overnight), inexpensive, simultaneous screening of several pathogens. The design of the antibody microarray is conducive to further development, and the panel of analyzed pathogens can be expanded to include approximately 50 members.
Collapse
Affiliation(s)
| | | | - Kirill I Lebedev
- Smorodintsev Research Institute of Influenza, St. Petersburg, Russia; Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| | - Alexey A Lozhkov
- Smorodintsev Research Institute of Influenza, St. Petersburg, Russia; Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Aleksandr S Taraskin
- Smorodintsev Research Institute of Influenza, St. Petersburg, Russia; Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Natalia E Gyulikhandanova
- Smorodintsev Research Institute of Influenza, St. Petersburg, Russia; Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Edward S Ramsay
- Smorodintsev Research Institute of Influenza, St. Petersburg, Russia
| | - Andrey V Vasin
- Smorodintsev Research Institute of Influenza, St. Petersburg, Russia; Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| |
Collapse
|
12
|
Soda K, Sakita M, Usui T, Ito H, Ito T. H5N8 avian influenza virus acquires enhanced pathogenicity after a single passage in chicken. Vet Microbiol 2019; 237:108381. [PMID: 31585646 DOI: 10.1016/j.vetmic.2019.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/26/2019] [Accepted: 08/06/2019] [Indexed: 12/22/2022]
Abstract
The H5N8 highly pathogenic avian influenza viruses (HPAIVs) isolated in Japan during the 2014-2015 winter differed in their pathogenicity in chickens. In the present study, we examined the possibility that a comparatively less pathogenic strain was first brought into the country by migratory birds, and then acquired enhanced pathogenicity by infecting chicken flocks. We showed that the A/tundra swan/Tottori/C6nk/2014 (H5N8) (Tottori P0) strain required 10 days to kill all chickens via the intranasal route. However, Tottori P1-B, a strain recovered from the brain of a chicken infected with parental Tottori P0, showed enhanced pathogenicity; Tottori P1-B replicated significantly in the lung and liver, and killed all infected birds within 6 days, which was comparable to a chicken farm isolate obtained in the same season, A/environment/Miyazaki/11/2014 (H5N8). Tottori P1-B showed more marked proliferation in MDCK and chicken fibroblast cells, especially during the early phase of infection. Sequence analysis revealed a single mutation, M374 V, in nucleoprotein (NP) of the passaged virus, and this substitution was conserved after a further inoculation study. Position 374 in NP is located in the functional domain interacting with polymerase protein, PB2, indicating that viral polymerase activity was involved in the rapid growth of Tottori P1-B in vitro and in vivo. These results suggest that HPAIV, which originally had comparatively low pathogenicity to chickens, can increase its pathogenicity through the infection from migratory birds to domestic chickens.
Collapse
Affiliation(s)
- Kosuke Soda
- Department of Joint Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan; Avian Zoonosis Research Center, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
| | - Masakazu Sakita
- Department of Joint Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
| | - Tatsufumi Usui
- Department of Joint Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan; Avian Zoonosis Research Center, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
| | - Hiroshi Ito
- Department of Joint Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan; Avian Zoonosis Research Center, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
| | - Toshihiro Ito
- Department of Joint Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan; Avian Zoonosis Research Center, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan.
| |
Collapse
|
13
|
Martínez-Sobrido L, Peersen O, Nogales A. Temperature Sensitive Mutations in Influenza A Viral Ribonucleoprotein Complex Responsible for the Attenuation of the Live Attenuated Influenza Vaccine. Viruses 2018; 10:E560. [PMID: 30326610 PMCID: PMC6213772 DOI: 10.3390/v10100560] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/03/2018] [Accepted: 10/12/2018] [Indexed: 01/29/2023] Open
Abstract
Live attenuated influenza vaccines (LAIV) have prevented morbidity and mortality associated with influenza viral infections for many years and represent the best therapeutic option to protect against influenza viral infections in humans. However, the development of LAIV has traditionally relied on empirical methods, such as the adaptation of viruses to replicate at low temperatures. These approaches require an extensive investment of time and resources before identifying potential vaccine candidates that can be safely implemented as LAIV to protect humans. In addition, the mechanism of attenuation of these vaccines is poorly understood in some cases. Importantly, LAIV are more efficacious than inactivated vaccines because their ability to mount efficient innate and adaptive humoral and cellular immune responses. Therefore, the design of potential LAIV based on known properties of viral proteins appears to be a highly appropriate option for the treatment of influenza viral infections. For that, the viral RNA synthesis machinery has been a research focus to identify key amino acid substitutions that can lead to viral attenuation and their use in safe, immunogenic, and protective LAIV. In this review, we discuss the potential to manipulate the influenza viral RNA-dependent RNA polymerase (RdRp) complex to generate attenuated forms of the virus that can be used as LAIV for the treatment of influenza viral infections, one of the current and most effective prophylactic options for the control of influenza in humans.
Collapse
Affiliation(s)
- Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York, NY 14642, USA.
| | - Olve Peersen
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, CO 80523, USA.
| | - Aitor Nogales
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York, NY 14642, USA.
| |
Collapse
|
14
|
Abstract
Influenza is a negative-sense single-stranded RNA virus with segmented genome. Each segment is encapsidated by a ribonucleoprotein (RNP) complex composed of RNA-dependent RNA polymerase (RdRP) and multiple copies of nucleoprotein (NP). The RNP complex plays a crucial role in viral life cycle, supporting and regulating transcription and replication of viral genome in infected cells. The structural characterization of RdRP and RNP in recent years has shed light on its functions and mechanism of action. In this review, we summarize current understanding on the structure of RNP complex, as well as the structure of each subunit. Crucial functions of RNP are also discussed.
Collapse
Affiliation(s)
- Chun-Yeung Lo
- Centre for Protein Science and Crystallography, School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, China
| | - Yun-Sang Tang
- Centre for Protein Science and Crystallography, School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, China
| | - Pang-Chui Shaw
- Centre for Protein Science and Crystallography, School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong, China.
| |
Collapse
|
15
|
Chen L, Wang C, Luo J, Li M, Liu H, Zhao N, Huang J, Zhu X, Ma G, Yuan G, He H. Amino Acid Substitution K470R in the Nucleoprotein Increases the Virulence of H5N1 Influenza A Virus in Mammals. Front Microbiol 2017; 8:1308. [PMID: 28744280 PMCID: PMC5504190 DOI: 10.3389/fmicb.2017.01308] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/28/2017] [Indexed: 12/21/2022] Open
Abstract
H5N1 is a highly pathogenic influenza A virus (IAV) and poses a major threat to the public health. The nucleoprotein (NP) has a multiple functions during the viral life cycle, however, the precise role of NP mutants in viral replication and pathogenicity is not completely understood. Here, we attempted to identify five residues in NP that may contribute to viral replication or pathogenicity. Of these, K227R, K229R, and K470R viruses were successfully rescued by reverse genetic, but the K91R and K198R viruses were not viable. A mini-genome assay demonstrated that the NP mutations K91R and K198R significantly decreased the polymerase activity. Moreover, these two mutations resulted in disrupted cellular localization in mammalian cells. Importantly, mutation at position 470 of NP significantly increased its virulence in vitro and in vivo. These findings demonstrated that the NP protein plays a major role in influenza virulence and pathogenicity, which adds to the knowledge of IAV virulence determinants and may benefit IAV surveillance.
Collapse
Affiliation(s)
- Lin Chen
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of SciencesBeijing, China.,University of the Chinese Academy of SciencesBeijing, China
| | - Chengmin Wang
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of SciencesBeijing, China
| | - Jing Luo
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of SciencesBeijing, China
| | - Meng Li
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of SciencesBeijing, China.,University of the Chinese Academy of SciencesBeijing, China
| | - Huimin Liu
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of SciencesBeijing, China.,University of the Chinese Academy of SciencesBeijing, China
| | - Na Zhao
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of SciencesBeijing, China.,University of the Chinese Academy of SciencesBeijing, China
| | - Jingjing Huang
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of SciencesBeijing, China
| | - Xili Zhu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of ScienceBeijing, China
| | - Guoyao Ma
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of SciencesBeijing, China.,University of the Chinese Academy of SciencesBeijing, China
| | - Guohui Yuan
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of SciencesBeijing, China
| | - Hongxuan He
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of SciencesBeijing, China
| |
Collapse
|
16
|
Liu Q, Liu Y, Yang J, Huang X, Han K, Zhao D, Bi K, Li Y. Two Genetically Similar H9N2 Influenza A Viruses Show Different Pathogenicity in Mice. Front Microbiol 2016; 7:1737. [PMID: 27867373 PMCID: PMC5096341 DOI: 10.3389/fmicb.2016.01737] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/17/2016] [Indexed: 12/02/2022] Open
Abstract
H9N2 Avian influenza virus has repeatedly infected humans and other mammals, which highlights the need to determine the pathogenicity and the corresponding mechanism of this virus for mammals. In this study, we found two H9N2 viruses with similar genetic background but with different pathogenicity in mice. The A/duck/Nanjing/06/2003 (NJ06) virus was highly pathogenic for mice, with a 50% mouse lethal dose (MLD50) of 102.83 50% egg infectious dose (EID50), whereas the A/duck/Nanjing/01/1999 (NJ01) virus was low pathogenic for mice, with a MLD50 of >106.81 EID50. Further studies showed that the NJ06 virus grew faster and reached significantly higher titers than NJ01 in vivo and in vitro. Moreover, the NJ06 virus induced more severe lung lesions, and higher levels of inflammatory cellular infiltration and cytokine response in lungs than NJ01 did. However, only 12 different amino acid residues (HA-K157E, NA-A9T, NA-R435K, PB2-T149P, PB2-K627E, PB1-R187K, PA-L548M, PA-M550L, NP-G127E, NP-P277H, NP-D340N, NS1-D171N) were found between the two viruses, and all these residues except for NA-R435K were located in the known functional regions involved in interaction of viral proteins or between the virus and host factors. Summary, our results suggest that multiple amino acid differences may be responsible for the higher pathogenicity of the NJ06 virus for mice, resulting in lethal infection, enhanced viral replication, severe lung lesions, and excessive inflammatory cellular infiltration and cytokine response in lungs. These observations will be helpful for better understanding the pathogenic potential and the corresponding molecular basis of H9N2 viruses that might pose threats to human health in the future.
Collapse
Affiliation(s)
- Qingtao Liu
- Key Laboratory of Veterinary Biological Engineering and Technology, National Center for Engineering Research of Veterinary Bio-products, Institute of Veterinary Medicine, Ministry of Agriculture, Jiangsu Academy of Agricultural SciencesNanjing, China; Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou, China
| | - Yuzhuo Liu
- Key Laboratory of Veterinary Biological Engineering and Technology, National Center for Engineering Research of Veterinary Bio-products, Institute of Veterinary Medicine, Ministry of Agriculture, Jiangsu Academy of Agricultural SciencesNanjing, China; Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou, China
| | - Jing Yang
- Key Laboratory of Veterinary Biological Engineering and Technology, National Center for Engineering Research of Veterinary Bio-products, Institute of Veterinary Medicine, Ministry of Agriculture, Jiangsu Academy of Agricultural SciencesNanjing, China; Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou, China
| | - Xinmei Huang
- Key Laboratory of Veterinary Biological Engineering and Technology, National Center for Engineering Research of Veterinary Bio-products, Institute of Veterinary Medicine, Ministry of Agriculture, Jiangsu Academy of Agricultural SciencesNanjing, China; Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou, China
| | - Kaikai Han
- Key Laboratory of Veterinary Biological Engineering and Technology, National Center for Engineering Research of Veterinary Bio-products, Institute of Veterinary Medicine, Ministry of Agriculture, Jiangsu Academy of Agricultural SciencesNanjing, China; Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou, China
| | - Dongmin Zhao
- Key Laboratory of Veterinary Biological Engineering and Technology, National Center for Engineering Research of Veterinary Bio-products, Institute of Veterinary Medicine, Ministry of Agriculture, Jiangsu Academy of Agricultural SciencesNanjing, China; Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou, China
| | - Keran Bi
- Key Laboratory of Veterinary Biological Engineering and Technology, National Center for Engineering Research of Veterinary Bio-products, Institute of Veterinary Medicine, Ministry of Agriculture, Jiangsu Academy of Agricultural SciencesNanjing, China; Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou, China
| | - Yin Li
- Key Laboratory of Veterinary Biological Engineering and Technology, National Center for Engineering Research of Veterinary Bio-products, Institute of Veterinary Medicine, Ministry of Agriculture, Jiangsu Academy of Agricultural SciencesNanjing, China; Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhou, China
| |
Collapse
|
17
|
Intrinsically disordered region of influenza A NP regulates viral genome packaging via interactions with viral RNA and host PI(4,5)P2. Virology 2016; 496:116-126. [PMID: 27289560 DOI: 10.1016/j.virol.2016.05.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/20/2016] [Accepted: 05/23/2016] [Indexed: 10/21/2022]
Abstract
To be incorporated into progeny virions, the viral genome must be transported to the inner leaflet of the plasma membrane (PM) and accumulate there. Some viruses utilize lipid components to assemble at the PM. For example, simian virus 40 (SV40) targets the ganglioside GM1 and human immunodeficiency virus type 1 (HIV-1) utilizes phosphatidylinositol (4,5) bisphosphate [PI(4,5)P2]. Recent studies clearly indicate that Rab11-mediated recycling endosomes are required for influenza A virus (IAV) trafficking of vRNPs to the PM but it remains unclear how IAV vRNP localized or accumulate underneath the PM for viral genome incorporation into progeny virions. In this study, we found that the second intrinsically disordered region (IDR2) of NP regulates two binding steps involved in viral genome packaging. First, IDR2 facilitates NP oligomer binding to viral RNA to form vRNP. Secondly, vRNP assemble by interacting with PI(4,5)P2 at the PM via IDR2. These findings suggest that PI(4,5)P2 functions as the determinant of vRNP accumulation at the PM.
Collapse
|
18
|
Liu CL, Hung HC, Lo SC, Chiang CH, Chen IJ, Hsu JTA, Hou MH. Using mutagenesis to explore conserved residues in the RNA-binding groove of influenza A virus nucleoprotein for antiviral drug development. Sci Rep 2016; 6:21662. [PMID: 26916998 PMCID: PMC4768256 DOI: 10.1038/srep21662] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 01/14/2016] [Indexed: 11/09/2022] Open
Abstract
Nucleoprotein (NP) is the most abundant type of RNA-binding viral protein in influenza A virus-infected cells and is necessary for viral RNA transcription and replication. Recent studies demonstrated that influenza NP is a valid target for antiviral drug development. The surface of the groove, covered with numerous conserved residues between the head and body domains of influenza A NP, plays a crucial role in RNA binding. To explore the mechanism by which NP binds RNA, we performed a series of site-directed mutagenesis in the RNA-binding groove, followed by surface plasmon resonance (SPR), to characterize the interactions between RNA and NP. Furthermore, a role of Y148 in NP stability and NP-RNA binding was evaluated. The aromatic residue of Y148 was found to stack with a nucleotide base. By interrupting the stacking interaction between Y148 and an RNA base, we identified an influenza virus NP inhibitor, (E, E)-1,7-bis(4-hydroxy-3-methoxyphenyl) -1,6-heptadiene-3,5-dione; this inhibitor reduced the NP's RNA-binding affinity and hindered viral replication. Our findings will be useful for the development of new drugs that disrupt the interaction between RNA and viral NP in the influenza virus.
Collapse
Affiliation(s)
- Chia-Lin Liu
- National Chung Hsing University, Department of Life Science, Taichung, 40227, Taiwan
| | - Hui-Chen Hung
- National Health Research Institutes, Institute of Biotechnology and Pharmaceutical Research, Miaoli, 35053, Taiwan
| | - Shou-Chen Lo
- National Chung Hsing University, Institute of Genomics and Bioinformatics, Taichung, 40227, Taiwan
| | - Ching-Hui Chiang
- National Chung Hsing University, Department of Life Science, Taichung, 40227, Taiwan
| | - I-Jung Chen
- National Health Research Institutes, Institute of Biotechnology and Pharmaceutical Research, Miaoli, 35053, Taiwan
| | - John T-A Hsu
- National Health Research Institutes, Institute of Biotechnology and Pharmaceutical Research, Miaoli, 35053, Taiwan
| | - Ming-Hon Hou
- National Chung Hsing University, Department of Life Science, Taichung, 40227, Taiwan.,National Chung Hsing University, Institute of Genomics and Bioinformatics, Taichung, 40227, Taiwan.,National Chung Hsing University, Institute of Biotechnology, Taichung, 40227, Taiwan
| |
Collapse
|
19
|
Li Q, Wang X, Sun Z, Hu J, Gao Z, Hao X, Li J, Liu H, Wang X, Gu M, Xu X, Liu X, Liu X. Adaptive mutations in PB2 gene contribute to the high virulence of a natural reassortant H5N2 avian influenza virus in mice. Virus Res 2015; 210:255-63. [PMID: 26315686 DOI: 10.1016/j.virusres.2015.08.017] [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: 03/10/2015] [Revised: 08/06/2015] [Accepted: 08/21/2015] [Indexed: 01/22/2023]
Abstract
The highly pathogenic A/chicken/Hebei/1102/2010 (HB10) H5N2 virus is a natural reassortant derived from circulating H5N1 and endemic H9N2 avian influenza viruses (AIV). To evaluate the potential of its interspecies transmission, we previously serially passaged the non-virulent HB10 virus in the mouse lung and obtained a high virulence variant (HB10-MA). Genomic sequencing revealed five mutations (HA-S227N, PB2-Q591K, PB2-D701N, PA-I554V and NP-R351K) that distinguished HB10-MA virus from its parental HB10 virus. In this study, we further investigated the molecular basis for the enhanced virulence of HB10-MA in mice. By generating a series of reassortants between the two viruses and evaluating their virulence in mice, we found that both PB2 and PA genes contribute to the high virulence of HB10-MA in mice, whereas PB2 gene carrying the 591K and/or 701N had a dominant function. In addition, the two amino acids showed a cumulative effect on the virulence, virus replication, and polymerase activity of HB10 or HB10-MA. Therefore, our results collectively emphasized the crucial role of PB2 gene, particularly the paired mutations of Q591K and D701N in the host adaptation of the novel reassortant H5N2 AIV in mammals, which may provide helpful insights into the pathogenic potential of emerging AIV in human beings.
Collapse
Affiliation(s)
- Qunhui Li
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xuan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Zhongtao Sun
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Jiao Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Zhao Gao
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xiaoli Hao
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Juan Li
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Huimou Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Min Gu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xiulong Xu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China.
| | - Xiaowen Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
20
|
Tarus B, Bertrand H, Zedda G, Di Primo C, Quideau S, Slama-Schwok A. Structure-based design of novel naproxen derivatives targeting monomeric nucleoprotein of Influenza A virus. J Biomol Struct Dyn 2014; 33:1899-912. [PMID: 25333630 PMCID: PMC4548311 DOI: 10.1080/07391102.2014.979230] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The nucleoprotein (NP) binds the viral RNA genome as oligomers assembled with the polymerase in a ribonucleoprotein complex required for transcription and replication of influenza A virus. Novel antiviral candidates targeting the nucleoprotein either induced higher order oligomers or reduced NP oligomerization by targeting the oligomerization loop and blocking its insertion into adjacent nucleoprotein subunit. In this study, we used a different structure-based approach to stabilize monomers of the nucleoprotein by drugs binding in its RNA-binding groove. We recently identified naproxen as a drug competing with RNA binding to NP with antiinflammatory and antiviral effects against influenza A virus. Here, we designed novel derivatives of naproxen by fragment extension for improved binding to NP. Molecular dynamics simulations suggested that among these derivatives, naproxen A and C0 were most promising. Their chemical synthesis is described. Both derivatives markedly stabilized NP monomer against thermal denaturation. Naproxen C0 bound tighter to NP than naproxen at a binding site predicted by MD simulations and shown by competition experiments using wt NP or single-point mutants as determined by surface plasmon resonance. MD simulations suggested that impeded oligomerization and stabilization of monomeric NP is likely to be achieved by drugs binding in the RNA grove and inducing close to their binding site conformational changes of key residues hosting the oligomerization loop as observed for the naproxen derivatives. Naproxen C0 is a potential antiviral candidate blocking influenza nucleoprotein function.
Collapse
Affiliation(s)
- Bogdan Tarus
- a Virologie et Immunologie Moléculaires, UR892, Institut National de la Recherche Agronomique , Domaine de Vilvert, 78350 Jouy en Josas , France
| | | | | | | | | | | |
Collapse
|
21
|
Mutations to PB2 and NP proteins of an avian influenza virus combine to confer efficient growth in primary human respiratory cells. J Virol 2014; 88:13436-46. [PMID: 25210184 DOI: 10.1128/jvi.01093-14] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Influenza pandemics occur when influenza A viruses (IAV) adapted to other host species enter humans and spread through the population. Pandemics are relatively rare due to host restriction of IAV: strains adapted to nonhuman species do not readily infect, replicate in, or transmit among humans. IAV can overcome host restriction through reassortment or adaptive evolution, and these are mechanisms by which pandemic strains arise in nature. To identify mutations that facilitate growth of avian IAV in humans, we have adapted influenza A/duck/Alberta/35/1976 (H1N1) (dk/AB/76) virus to a high-growth phenotype in differentiated human tracheo-bronchial epithelial (HTBE) cells. Following 10 serial passages of three independent lineages, the bulk populations showed similar growth in HTBE cells to that of a human seasonal virus. The coding changes present in six clonal isolates were determined. The majority of changes were located in the polymerase complex and nucleoprotein (NP), and all isolates carried mutations in the PB2 627 domain and regions of NP thought to interact with PB2. Using reverse genetics, the impact on growth and polymerase activity of individual and paired mutations in PB2 and NP was evaluated. The results indicate that coupling of the mammalian-adaptive mutation PB2 E627K or Q591K to selected mutations in NP further augments the growth of the corresponding viruses. In addition, minimal combinations of three (PB2 Q236H, E627K, and NP N309K) or two (PB2 Q591K and NP S50G) mutations were sufficient to recapitulate the efficient growth in HTBE cells of dk/AB/76 viruses isolated after 10 passages in this substrate. IMPORTANCE Influenza A viruses adapted to birds do not typically grow well in humans. However, as has been seen recently with H5N1 and H7N9 subtype viruses, productive and virulent infection of humans with avian influenza viruses can occur. The ability of avian influenza viruses to adapt to new host species is a consequence of their high mutation rate that supports their zoonotic potential. Understanding of the adaptation of avian viruses to mammals strengthens public health efforts aimed at controlling influenza. In particular, it is critical to know how readily and through mutation to which functional components avian influenza viruses gain the ability to grow efficiently in humans. Our data show that as few as three mutations, in the PB2 and NP proteins, support robust growth of a low-pathogenic, H1N1 duck isolate in primary human respiratory cells.
Collapse
|
22
|
Abstract
The influenza A virus causes a highly contagious respiratory disease that significantly impacts our economy and health. Its replication and transcription is catalyzed by the viral RNA polymerase. This enzyme is also crucial for the virus, because it is involved in the adaptation of zoonotic strains. It is thus of major interest for the development of antiviral therapies and is being intensively studied. In this article, we will discuss recent advances that have improved our knowledge of the structure of the RNA polymerase and how mutations in the polymerase help the virus to spread effectively among new hosts.
Collapse
Affiliation(s)
- Thomas M Stubbs
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK ; Babraham Institute, Brabraham Research Campus, Cambridge, CB22 3AT, UK
| | - Aartjan Jw Te Velthuis
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| |
Collapse
|
23
|
Gui X, Li R, Zhang X, Shen C, Yu H, Guo X, Kang Y, Chen J, Chen H, Chen Y, Xia N. An important amino acid in nucleoprotein contributes to influenza A virus replication by interacting with polymerase PB2. Virology 2014; 464-465:11-20. [PMID: 25043584 DOI: 10.1016/j.virol.2014.06.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 05/07/2014] [Accepted: 06/24/2014] [Indexed: 11/24/2022]
Abstract
The nucleoprotein (NP) of influenza A virus plays a critical role in the formation of viral ribonucleoprotein (vRNP) complex. However, it remains unclear which key residues in NP are associated with the assembly of vRNP and contribute to virus replication. Here, a highly conserved aspartic acid at residue 88 (D88) of NP was identified by molecular docking of NP with the Fv region of a broad-spectrum anti-NP mAb 19C10 and further demonstrated to be an important residue contributes to the RNP activity, virus growth in MDCK cells and replication in lungs of infected mice by comparing recombinant wild-type A/WSN/1933 virus to the mutant virus that contains an alanine instead of aspartic acid at NP residue 88. D88 was also predicted to interact with PB2 by molecular docking and further verified by immunoprecipitation. These findings provide new information for understanding the interaction between NP and other polymerase subunits in virus replication.
Collapse
Affiliation(s)
- Xun Gui
- National Institute of Diagnostics and Vaccine Development in Infectiou Diseases, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Rui Li
- National Institute of Diagnostics and Vaccine Development in Infectiou Diseases, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, Fujian, China
| | - Xuhui Zhang
- National Institute of Diagnostics and Vaccine Development in Infectiou Diseases, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Chenguang Shen
- National Institute of Diagnostics and Vaccine Development in Infectiou Diseases, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Hai Yu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, Fujian, China
| | - Xiaoyi Guo
- National Institute of Diagnostics and Vaccine Development in Infectiou Diseases, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Yahong Kang
- National Institute of Diagnostics and Vaccine Development in Infectiou Diseases, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Junyu Chen
- National Institute of Diagnostics and Vaccine Development in Infectiou Diseases, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Honglin Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, Fujian, China; State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Hong Kong SAR, China
| | - Yixin Chen
- National Institute of Diagnostics and Vaccine Development in Infectiou Diseases, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, Fujian, China.
| | - Ningshao Xia
- National Institute of Diagnostics and Vaccine Development in Infectiou Diseases, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, Fujian, China.
| |
Collapse
|
24
|
Liu Q, Chen H, Huang J, Chen Y, Gu M, Wang X, Hu S, Liu X, Liu X. A nonpathogenic duck-origin H9N2 influenza A virus adapts to high pathogenicity in mice. Arch Virol 2014; 159:2243-52. [PMID: 24696271 DOI: 10.1007/s00705-014-2062-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 03/19/2014] [Indexed: 11/26/2022]
Abstract
H9N2 influenza viruses continue to circulate in wild birds and poultry in Eurasian countries and have repeatedly infected mammals, including pigs and humans, posing a significant threat to public health. To understand the adaptation of H9N2 influenza viruses to mammals, we serially passaged a nonpathogenic duck-origin H9N2 influenza virus, A/duck/Jiangsu/1/2008 (DK1), in mouse lungs. Increased virulence was detectable after five sequential passages, and a highly pathogenic mouse-adapted strain (DK1-MA) with a 50% mouse lethal dose of 10(2.37) 50% egg infectious dose was obtained after 18 passages. DK1-MA grew faster and reached significantly higher titers than DK1 in mouse lungs and could sporadically spread to other organs. Moreover, DK1-MA induced a greater magnitude of pulmonary edema and higher levels of inflammatory cellular infiltration in bronchoalveolar lavage fluids than DK1 did. Genomic sequence alignment revealed eight amino acid substitutions (HA-L80F, HA-N193D, NA-A27T, PB2-F404L, PA-D3V, PA-S225R, NP-V105M, M1-A166V) in six viral proteins of DK1-MA compared with DK1 virus. Except for HA-L80F, the other seven substitutions were all located in known functional regions involved in interaction of viral proteins or interaction between the virus and host factors. Taken together, our results suggest that multiple amino acid substitutions may be involved in the adaptation of H9N2 avian influenza virus to mice, resulting in lethal infection, enhanced viral replication, severe pulmonary edema, and excessive inflammatory cellular infiltration in lungs. These observations provide helpful insights into the pathogenic potential of H9N2 avian influenza viruses that could pose threats to human health in the future.
Collapse
Affiliation(s)
- Qingtao Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Large-scale analysis of influenza A virus nucleoprotein sequence conservation reveals potential drug-target sites. Virology 2014; 454-455:40-7. [PMID: 24725930 DOI: 10.1016/j.virol.2014.01.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/18/2013] [Accepted: 01/24/2014] [Indexed: 01/05/2023]
Abstract
The nucleoprotein (NP) of the influenza A virus encapsidates the viral RNA and participates in the infectious life cycle of the virus. The aims of this study were to find the degree of conservation of NP among all virus subtypes and hosts and to identify conserved binding sites, which may be utilised as potential drug target sites. The analysis of conservation based on 4430 amino acid sequences identified high conservation in known functional regions as well as novel highly conserved sites. Highly variable clusters identified on the surface of NP may be associated with adaptation to different hosts and avoidance of the host immune defence. Ligand binding potential overlapping with high conservation was found in the tail-loop binding site and near the putative RNA binding region. The results provide the basis for developing antivirals that may be universally effective and have a reduced potential to induce resistance through mutations.
Collapse
|
26
|
Structure-based discovery of the novel antiviral properties of naproxen against the nucleoprotein of influenza A virus. Antimicrob Agents Chemother 2013; 57:2231-42. [PMID: 23459490 DOI: 10.1128/aac.02335-12] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The nucleoprotein (NP) binds the viral RNA genome and associates with the polymerase in a ribonucleoprotein complex (RNP) required for transcription and replication of influenza A virus. NP has no cellular counterpart, and the NP sequence is highly conserved, which led to considering NP a hot target in the search for antivirals. We report here that monomeric nucleoprotein can be inhibited by a small molecule binding in its RNA binding groove, resulting in a novel antiviral against influenza A virus. We identified naproxen, an anti-inflammatory drug that targeted the nucleoprotein to inhibit NP-RNA association required for NP function, by virtual screening. Further docking and molecular dynamics (MD) simulations identified in the RNA groove two NP-naproxen complexes of similar levels of interaction energy. The predicted naproxen binding sites were tested using the Y148A, R152A, R355A, and R361A proteins carrying single-point mutations. Surface plasmon resonance, fluorescence, and other in vitro experiments supported the notion that naproxen binds at a site identified by MD simulations and showed that naproxen competed with RNA binding to wild-type (WT) NP and protected active monomers of the nucleoprotein against proteolytic cleavage. Naproxen protected Madin-Darby canine kidney (MDCK) cells against viral challenges with the H1N1 and H3N2 viral strains and was much more effective than other cyclooxygenase inhibitors in decreasing viral titers of MDCK cells. In a mouse model of intranasal infection, naproxen treatment decreased the viral titers in mice lungs. In conclusion, naproxen is a promising lead compound for novel antivirals against influenza A virus that targets the nucleoprotein in its RNA binding groove.
Collapse
|
27
|
Antony H, Schaeffer PM. A GFP-tagged nucleoprotein-based aggregation assay for anti-influenza drug discovery and antibody development. Analyst 2013; 138:6073-80. [DOI: 10.1039/c3an01041d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
28
|
Yamada K, Koyama H, Hagiwara K, Ueda A, Sasaki Y, Kanesashi SN, Ueno R, Nakamura HK, Kuwata K, Shimizu K, Suzuki M, Aida Y. Identification of a novel compound with antiviral activity against influenza A virus depending on PA subunit of viral RNA polymerase. Microbes Infect 2012; 14:740-7. [PMID: 22441116 DOI: 10.1016/j.micinf.2012.02.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 02/27/2012] [Accepted: 02/29/2012] [Indexed: 10/28/2022]
Abstract
Influenza viruses have developed resistance to current drugs, creating a need for new antiviral targets and new drugs to treat influenza virus infections. In this study, computational and experimental screening of an extensive compound library identified THC19, which was able to suppress influenza virus replication. This compound had no cytotoxic effects and did not disrupt cell cycle progression or induce apoptosis in MDCK cells as confirmed by WST-1 assays, flow cytometry analysis, and caspase-3 assays. Time-of-addition experiments showed that THC19 acts at a relatively early stage of the viral lifecycle. Subsequent mini-genome assays revealed that THC19 inhibited viral genome replication and/or transcription, suggesting that it interferes with one or more of the viral components that form the ribonucleoprotein complexes, namely polymerase basic 2 (PB2), polymerase basic 1 (PB1), polymerase acidic (PA), nucleoprotein (NP) and viral RNA. Finally, mini-genome assays where PB2, PB1, PA or NP from A/WSN/33 (H1N1) virus were replaced with those from A/Udorn/307/1972 (H3N2) virus effectively demonstrated that THC19 inhibited viral multiplication in a manner dependent upon the PA subunit. Taken together, these results suggest that influenza virus PA protein is a potential target for, and may aid the development of, novel compounds that inhibit influenza A virus replication.
Collapse
Affiliation(s)
- Kazunori Yamada
- Viral Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Cheng H, Wan J, Lin MI, Liu Y, Lu X, Liu J, Xu Y, Chen J, Tu Z, Cheng YSE, Ding K. Design, synthesis, and in vitro biological evaluation of 1H-1,2,3-triazole-4-carboxamide derivatives as new anti-influenza A agents targeting virus nucleoprotein. J Med Chem 2012; 55:2144-53. [PMID: 22332894 DOI: 10.1021/jm2013503] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The influenza virus nucleoprotein (NP) is an emerging target for anti-influenza drug development. Nucleozin (1) and its closely related derivatives had been identified as NP inhibitors displaying anti-influenza activity. Utilizing 1 as a lead molecule, we successfully designed and synthesized a series of 1H-1,2,3-triazole-4-carboxamide derivatives as new anti-influenza A agents. One of the most potent compounds, 3b, inhibited the replication of various H3N2 and H1N1 influenza A virus strains with IC(50) values ranging from 0.5 to 4.6 μM. Compound 3b also strongly inhibited the replication of H5N1 (RG14), amantidine-resistant A/WSN/33 (H1N1), and oseltamivir-resistant A/WSN/1933 (H1N1, 274Y) virus strains with IC(50) values in sub-μM ranges. Further computational studies and mechanism investigation suggested that 3b might directly target influenza virus A nucleoprotein to inhibit its nuclear accumulation.
Collapse
Affiliation(s)
- Huimin Cheng
- Key Laboratory of Regenerative Biology and Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Detection of highly pathogenic influenza and pandemic influenza virus in formalin fixed tissues by immunohistochemical methods. J Virol Methods 2012; 179:409-13. [DOI: 10.1016/j.jviromet.2011.11.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 10/29/2011] [Accepted: 11/09/2011] [Indexed: 11/23/2022]
|
31
|
Tarus B, Chevalier C, Richard CA, Delmas B, Di Primo C, Slama-Schwok A. Molecular dynamics studies of the nucleoprotein of influenza A virus: role of the protein flexibility in RNA binding. PLoS One 2012; 7:e30038. [PMID: 22272272 PMCID: PMC3260217 DOI: 10.1371/journal.pone.0030038] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 12/08/2011] [Indexed: 11/18/2022] Open
Abstract
The influenza viruses contain a segmented, negative stranded RNA genome. Each RNA segment is covered by multiple copies of the nucleoprotein (NP). X-ray structures have shown that NP contains well-structured domains juxtaposed with regions of missing electron densities corresponding to loops. In this study, we tested if these flexible loops gated or promoted RNA binding and RNA-induced oligomerization of NP. We first performed molecular dynamics simulations of wt NP monomer and trimer in comparison with the R361A protein mutated in the RNA binding groove, using the H1N1 NP as the initial structure. Calculation of the root-mean-square fluctuations highlighted the presence of two flexible loops in NP trimer: loop 1 (73-90), loop 2 (200-214). In NP, loops 1 and 2 formed a 10-15 Å-wide pinch giving access to the RNA binding groove. Loop 1 was stabilized by interactions with K113 of the adjacent β-sheet 1 (91-112) that interacted with the RNA grove (linker 360-373) via multiple hydrophobic contacts. In R361A, a salt bridge formed between E80 of loop 1 and R208 of loop 2 driven by hydrophobic contacts between L79 and W207, due to a decreased flexibility of loop 2 and loop 1 unfolding. Thus, RNA could not access its binding groove in R361A; accordingly, R361A had a much lower affinity for RNA than NP. Disruption of the E80-R208 interaction in the triple mutant R361A-E80A-E81A increased its RNA binding affinity and restored its oligomerization back to wt levels in contrast with impaired levels of R361A. Our data suggest that the flexibility of loops 1 and 2 is required for RNA sampling and binding which likely involve conformational change(s) of the nucleoprotein.
Collapse
Affiliation(s)
- Bogdan Tarus
- Virologie et Immunologie Moléculaires, UR 892, Centre INRA de Jouy en Josas, France
| | - Christophe Chevalier
- Virologie et Immunologie Moléculaires, UR 892, Centre INRA de Jouy en Josas, France
| | | | - Bernard Delmas
- Virologie et Immunologie Moléculaires, UR 892, Centre INRA de Jouy en Josas, France
| | - Carmelo Di Primo
- Université of Bordeaux, ARNA Laboratory, Bordeaux, France
- INSERM U869, ARNA Laboratory, Institut Européen de Chimie et de Biologie, Pessac, France
| | - Anny Slama-Schwok
- Virologie et Immunologie Moléculaires, UR 892, Centre INRA de Jouy en Josas, France
| |
Collapse
|
32
|
Combining RNA–DNA swapping and quantitative polymerase chain reaction for the detection of influenza A nucleoprotein. Anal Biochem 2012; 420:121-6. [DOI: 10.1016/j.ab.2011.09.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 09/08/2011] [Accepted: 09/09/2011] [Indexed: 12/25/2022]
|
33
|
Aggarwal S, Dewhurst S, Takimoto T, Kim B. Biochemical impact of the host adaptation-associated PB2 E627K mutation on the temperature-dependent RNA synthesis kinetics of influenza A virus polymerase complex. J Biol Chem 2011; 286:34504-13. [PMID: 21816827 DOI: 10.1074/jbc.m111.262048] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Most avian influenza A viruses, which preferentially replicate at the high temperatures found in the digestive tract of birds, have a glutamic acid at residue 627 of the viral RNA polymerase PB2 subunit (Glu-627), whereas the human viruses, which optimally replicate at the low temperatures observed in the human respiratory tract, have a lysine (Lys-627). The mechanism of action for this mutation is still not understood, although interaction with host factors has been proposed to play a major role. In this study, we explored an alternative, yet related, hypothesis that this PB2 mutation may alter the temperature-dependent enzymatic polymerase activity of the viral polymerase. First, the avian polymerase protein, which was purified from baculovirus expression system, indeed remained significantly active at higher temperatures (i.e. 37 and 42 °C), whereas the human E627K mutant drastically lost activity at these high temperatures. Second, our steady-state kinetics data revealed that the human E627K mutant polymerase is catalytically more active than the avian Glu-627 polymerase at 34 °C. Importantly, the E627K mutation elevates apparent K(cat) at low temperatures with little effect on K(m), suggesting that the E627K mutation alters the biochemical steps involved in enzyme catalysis rather than the interaction with the incoming NTP. Third, this temperature-dependent kinetic impact of the human E627K mutation was also observed with different RNA templates, with different primers and also in the presence of nucleoprotein. In conclusion, our study suggests that the amino acid sequence variations at residue 627 of PB2 subunit can directly alter the enzyme kinetics of influenza polymerase.
Collapse
Affiliation(s)
- Shilpa Aggarwal
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | |
Collapse
|
34
|
Sharma K, Tripathi S, Ranjan P, Kumar P, Garten R, Deyde V, Katz JM, Cox NJ, Lal RB, Sambhara S, Lal SK. Influenza A virus nucleoprotein exploits Hsp40 to inhibit PKR activation. PLoS One 2011; 6:e20215. [PMID: 21698289 PMCID: PMC3115951 DOI: 10.1371/journal.pone.0020215] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 04/15/2011] [Indexed: 02/08/2023] Open
Abstract
Background Double-stranded RNA dependent protein kinase (PKR) is a key regulator of the anti-viral innate immune response in mammalian cells. PKR activity is regulated by a 58 kilo Dalton cellular inhibitor (P58IPK), which is present in inactive state as a complex with Hsp40 under normal conditions. In case of influenza A virus (IAV) infection, P58IPK is known to dissociate from Hsp40 and inhibit PKR activation. However the influenza virus component responsible for PKR inhibition through P58IPK activation was hitherto unknown. Principal Findings Human heat shock 40 protein (Hsp40) was identified as an interacting partner of Influenza A virus nucleoprotein (IAV NP) using a yeast two-hybrid screen. This interaction was confirmed by co-immunoprecipitation studies from mammalian cells transfected with IAV NP expressing plasmid. Further, the IAV NP-Hsp40 interaction was validated in mammalian cells infected with various seasonal and pandemic strains of influenza viruses. Cellular localization studies showed that NP and Hsp40 co-localize primarily in the nucleus. During IAV infection in mammalian cells, expression of NP coincided with the dissociation of P58IPK from Hsp40 and decrease PKR phosphorylation. We observed that, plasmid based expression of NP in mammalian cells leads to decrease in PKR phosphorylation. Furthermore, inhibition of NP expression during influenza virus replication led to PKR activation and concomitant increase in eIF2α phosphorylation. Inhibition of NP expression also led to reduced IRF3 phosphorylation, enhanced IFN β production and concomitant reduction of virus replication. Taken together our data suggest that NP is the viral factor responsible for P58IPK activation and subsequent inhibition of PKR-mediated host response during IAV infection. Significance Our findings demonstrate a novel role of IAV NP in inhibiting PKR-mediated anti-viral host response and help us understand P58IPK mediated inhibition of PKR activity during IAV infection.
Collapse
Affiliation(s)
- Kulbhushan Sharma
- Virology Group, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
| | - Shashank Tripathi
- Virology Group, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
| | - Priya Ranjan
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Purnima Kumar
- Virology Group, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
| | - Rebecca Garten
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Varough Deyde
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jacqueline M. Katz
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Nancy J. Cox
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Renu B. Lal
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Suryaprakash Sambhara
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Sunil K. Lal
- Virology Group, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
- * E-mail:
| |
Collapse
|
35
|
Yan SM, Wu G. Fitting evolutionary process of influenza A virus nucleoproteins using analytical solution of system of differential equations. Interdiscip Sci 2011; 3:128-137. [PMID: 21541842 DOI: 10.1007/s12539-011-0078-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Revised: 01/18/2010] [Accepted: 02/02/2010] [Indexed: 05/30/2023]
Abstract
Very recently we explored the possibility of using differential equations to describe the evolution of proteins. In this study we used the amino-acid pair predictability to quantify 1709 nucleoproteins of influenza A viruses isolated from 1918 to 2008 to represent their evolutionary process, thereafter we used the analytical solution of system of differential equations to fit the evolution of the nucleoprotein family. The results showed that the analytical solution could fit the nucleoprotein evolution and the obtained parameters were useful for timing of future mutations. Our approach provided a way to quantitatively analyze protein dynamics and evolution.
Collapse
Affiliation(s)
- Shao-Min Yan
- State Key Laboratory of Non-food Biomass Enzyme Technology, National Engineering Research Center for Non-food Biorefinery, Guangxi Key Laboratory of Biorefinery, Guangxi Academy of Sciences, 98 Daling Road, Nanning, Guangxi, 530007, China
| | | |
Collapse
|
36
|
Focused on Frontier Sciences and Dedicated to Academic Exchanges-<I>Science China: Life Sciences</I> in 2009 at a New Beginning. PROG BIOCHEM BIOPHYS 2011. [DOI: 10.3724/sp.j.1206.2010.00555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
37
|
Hua Q. Two interesting biological fields: Avian influenza virus and epigenetics. CHINESE SCIENCE BULLETIN-CHINESE 2010; 55:4094-4096. [PMID: 32214735 PMCID: PMC7088840 DOI: 10.1007/s11434-010-4239-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 10/22/2010] [Indexed: 11/29/2022]
Affiliation(s)
- QingXin Hua
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106-4935 USA
| |
Collapse
|
38
|
|
39
|
Research frontiers highlighted in Science China Life Sciences in 2009. CHINESE SCIENCE BULLETIN-CHINESE 2010. [DOI: 10.1007/s11434-010-4229-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
40
|
Hu Y. Two special topics on the avian influenza virus and on epigenetics, have drawn much attention. SCIENCE CHINA. LIFE SCIENCES 2010; 53:1483-1484. [PMID: 21181351 DOI: 10.1007/s11427-010-4105-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 11/10/2010] [Indexed: 05/30/2023]
Affiliation(s)
- Yonglin Hu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
41
|
Gao GF, Sun Y. It is not just AIV: from avian to swine-origin influenza virus. SCIENCE CHINA-LIFE SCIENCES 2010; 53:151-153. [PMID: 20596968 DOI: 10.1007/s11427-010-0017-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- George F Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China. .,Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, China.
| | - YePing Sun
- CAS Key Laboratory of Pathogenic Microbiology and Immunology Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
42
|
Yan S, Wu G. Trends in global warming and evolution of nucleoproteins from influenza A viruses since 1918. Transbound Emerg Dis 2010; 57:404-13. [PMID: 20825589 DOI: 10.1111/j.1865-1682.2010.01164.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Global warming affects not only the environment where we live, but also all living species to different degree, including influenza A virus. We recently conducted several studies on the possible impact of global warming on the protein families of influenza A virus. More studies are needed in order to have a full picture of the impact of global warming on living organisms, especially its effect on viruses. In this study, we correlate trends in global warming with evolution of the nucleoprotein from influenza A virus and then analyse the trends with respect to northern/southern hemispheres, virus subtypes and sampling species. The results suggest that global warming may have an impact on the evolution of the nucleoprotein from influenza A virus.
Collapse
Affiliation(s)
- S Yan
- State Key Laboratory of Non-food Biomass Enzyme Technology, National Engineering Research Center for Non-food Biorefinery, Guangxi Key Laboratory of Biorefinery, Guangxi Academy of Sciences, Nanning, Guangxi, China
| | | |
Collapse
|
43
|
Dillon D, Runstadler J. Mx gene diversity and influenza association among five wild dabbling duck species (Anas spp.) in Alaska. INFECTION GENETICS AND EVOLUTION 2010; 10:1085-93. [PMID: 20621205 DOI: 10.1016/j.meegid.2010.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 07/01/2010] [Accepted: 07/02/2010] [Indexed: 12/29/2022]
Abstract
Mx (myxovirus-resistant) proteins are induced by interferon and inhibit viral replication as part of the innate immune response to viral infection in many vertebrates. Influenza A virus appears to be especially susceptible to Mx antiviral effects. We characterized exon 13 and the 3' UTR of the Mx gene in wild ducks, the natural reservoir of influenza virus and explored its potential relevance to influenza infection. We observed a wide range of intra- and interspecies variations. Total nucleotide diversity per site was 0.0014, 0.0027, 0.0044, 0.0051, and 0.0061 in mallards, northern shovelers, northern pintails, American wigeon, and American green-winged teals, respectively. There were 61 haplotypes present across all five species and four were shared among species. Additionally, we observed a significant association between Mx haplotype and influenza infection status in northern shovelers. However, we found no evidence of balancing or diversifying selection in this region of the Mx gene. Characterization of the duck Mx gene is an important step in understanding how the gene may affect disease resistance or susceptibility in wild populations. Furthermore, given that waterfowl act as a natural reservoir for influenza virus, the Mx gene could be an important determinant in the ecology of the virus.
Collapse
Affiliation(s)
- Danielle Dillon
- Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, USA.
| | | |
Collapse
|
44
|
Abstract
The molecular mechanism by which pandemic 2009 influenza A viruses were able to sufficiently adapt to humans is largely unknown. Subsequent human infections with novel H1N1 influenza viruses prompted an investigation of the molecular determinants of the host range and pathogenicity of pandemic influenza viruses in mammals. To address this problem, we assessed the genetic basis for increased virulence of A/CA/04/09 (H1N1) and A/TN/1-560/09 (H1N1) isolates, which are not lethal for mice, in a new mammalian host by promoting their mouse adaptation. The resulting mouse lung-adapted variants showed significantly enhanced growth characteristics in eggs, extended extrapulmonary tissue tropism, and pathogenicity in mice. All mouse-adapted viruses except A/TN/1-560/09-MA2 grew faster and to higher titers in cells than the original strains. We found that 10 amino acid changes in the ribonucleoprotein (RNP) complex (PB2 E158G/A, PA L295P, NP D101G, and NP H289Y) and hemagglutinin (HA) glycoprotein (K119N, G155E, S183P, R221K, and D222G) controlled enhanced mouse virulence of pandemic isolates. HA mutations acquired during adaptation affected viral receptor specificity by enhancing binding to alpha2,3 together with decreasing binding to alpha2,6 sialyl receptors. PB2 E158G/A and PA L295P amino acid substitutions were responsible for the significant enhancement of transcription and replication activity of the mouse-adapted H1N1 variants. Taken together, our findings suggest that changes optimizing receptor specificity and interaction of viral polymerase components with host cellular factors are the major mechanisms that contribute to the optimal competitive advantage of pandemic influenza viruses in mice. These modulators of virulence, therefore, may have been the driving components of early evolution, which paved the way for novel 2009 viruses in mammals.
Collapse
|
45
|
Salahuddin P, Khan AU. Structure function studies on different structural domains of nucleoprotein of H1N1 subtype. Bioinformation 2010; 5:28-30. [PMID: 21346875 PMCID: PMC3040001 DOI: 10.6026/97320630005025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2010] [Accepted: 06/08/2010] [Indexed: 11/23/2022] Open
Abstract
Recent 2009 flu pandemic is a global outbreak of a new strain of influenza A virus subtype H1N1. The H1N1 virus has crossed species barrier to human
and apparently acquired the capability to transmit this disease from human to human. The NP is a multifunctional protein that not only encapsidates viral
RNA (vRNA), but also forms homo-oligomer and thereby maintains RNP structure. It is also thought to be the key adaptor for virus and host cell
interaction. Thus, it is one of the factor that play a key role in the pathogenesis of influenza A virus infection. Therefore, to understand the cause of
pathogenicity of H1N1 virus, we have studied the structure-function relationship of different domains of NP. Our results showed that conservative
mutation in NP of various strains were pathogenic in nature. However, non-conservative mutation slightly abrogated oligomerization and was therefore
less pathogenic. Our results also suggest that beside tail and body domain, head domain may also participate in an oligomerization process.
Collapse
Affiliation(s)
| | - Asad U Khan
- Distributed Information Sub-Centre
- Interdisciplinary Biotechnology Unit; A.M.U. Aligarh, 202002, India
- Asad U Khan Phone: +91 571 2723088; Fax: +91 571 2721776
| |
Collapse
|
46
|
Functional analysis of the influenza virus H5N1 nucleoprotein tail loop reveals amino acids that are crucial for oligomerization and ribonucleoprotein activities. J Virol 2010; 84:7337-45. [PMID: 20463064 DOI: 10.1128/jvi.02474-09] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Homo-oligomerization of the nucleoprotein (NP) of influenza A virus is crucial for providing a major structural framework for the assembly of viral ribonucleoprotein (RNP) particles. The nucleoprotein is also essential for transcription and replication during the virus life cycle. In the H5N1 NP structure, the tail loop region is important for NP to form oligomers. Here, by an RNP reconstitution assay, we identified eight NP mutants that had different degrees of defects in forming functional RNPs, with the RNP activities of four mutants being totally abolished (E339A, V408S P410S, R416A, and L418S P419S mutants) and the RNP activities of the other four mutants being more than 50% decreased (R267A, I406S, R422A, and E449A mutants). Further characterization by static light scattering showed that the totally defective protein variants existed as monomers in vitro, deviating from the trimeric/oligomeric form of wild-type NP. The I406S, R422A, and E449A variants existed as a mixture of unstable oligomers, thus resulting in a reduction of RNP activity. Although the R267A variant existed as a monomer in vitro, it resumed an oligomeric form upon the addition of RNA and retained a certain degree of RNP activity. Our data suggest that there are three factors that govern the NP oligomerization event: (i) interaction between the tail loop and the insertion groove, (ii) maintenance of the tail loop conformation, and (iii) stabilization of the NP homo-oligomer. The work presented here provides information for the design of NP inhibitors for combating influenza virus infection.
Collapse
|
47
|
Hagiwara K, Kondoh Y, Ueda A, Yamada K, Goto H, Watanabe T, Nakata T, Osada H, Aida Y. Discovery of novel antiviral agents directed against the influenza A virus nucleoprotein using photo-cross-linked chemical arrays. Biochem Biophys Res Commun 2010; 394:721-7. [DOI: 10.1016/j.bbrc.2010.03.058] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 03/09/2010] [Indexed: 01/17/2023]
|