1
|
Bernardo-Menezes LC, Agrelli A, Oliveira ASLED, Azevedo EDAN, Morais CNLD. Zika virus: Critical crosstalk between pathogenesis, cytopathic effects, and macroautophagy. J Cell Biochem 2024; 125:e30438. [PMID: 37334850 DOI: 10.1002/jcb.30438] [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: 02/23/2023] [Revised: 05/06/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023]
Abstract
Zika virus (ZIKV) is a re-emerging positive-sense RNA arbovirus. Its genome encodes a polyprotein that is cleaved by proteases into three structural proteins (Envelope, pre-Membrane, and Capsid) and seven nonstructural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5). These proteins have essential functions in viral replication cycle, cytopathic effects, and host cellular response. When infected by ZIKV, host cells promote macroautophagy, which is believed to favor virus entry. Although several authors have attempted to understand this link between macroautophagy and viral infection, little is known. Herein, we performed a narrative review of the molecular connection between macroautophagy and ZIKV infection while focusing on the roles of the structural and nonstructural proteins. We concluded that ZIKV proteins are major virulence factors that modulate host-cell machinery to its advantage by disrupting and/or blocking specific cellular systems and organelles' function, such as endoplasmic reticulum stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Lucas Coêlho Bernardo-Menezes
- Laboratory of Virology and Experimental Therapeutics (LaViTE), Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Almerinda Agrelli
- Laboratory of Nanostructured Materials (LMNANO), Strategic Technologies Center of Northeast (CETENE), Recife, Pernambuco, Brazil
| | | | - Elisa de Almeida Neves Azevedo
- Laboratory of Virology and Experimental Therapeutics (LaViTE), Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Clarice Neuenschwander Lins de Morais
- Laboratory of Virology and Experimental Therapeutics (LaViTE), Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| |
Collapse
|
2
|
Pan Q, Chen Q, Zhang W, Jiao H, Yu L, Hu H. Structural Insights into the Dynamic Assembly of a YFV sNS1 Tetramer. Viruses 2024; 16:1212. [PMID: 39205186 PMCID: PMC11359903 DOI: 10.3390/v16081212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 09/04/2024] Open
Abstract
Yellow fever virus (YFV) infections can cause severe diseases in humans, resulting in mass casualties in Africa and the Americas each year. Secretory NS1 (sNS1) is thought to be used as a diagnostic marker of flavivirus infections, playing an essential role in the flavivirus life cycle, but little is known about the composition and structure of YFV sNS1. Here, we present that the recombinant YFV sNS1 exists in a heterogeneous mixture of oligomerizations, predominantly in the tetrameric form. The cryoEM structures show that the YFV tetramer of sNS1 is stacked by the hydrophobic interaction between β-roll domains and greasy fingers. According to the 3D variability analysis, the tetramer is in a semi-stable state that may contain multiple conformations with dynamic changes. We believe that our study provides critical insights into the oligomerization of NS1 and will aid the development of NS1-based diagnoses and therapies.
Collapse
Affiliation(s)
- Qi Pan
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| | - Qiang Chen
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| | - Wanqin Zhang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| | - Haizhan Jiao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| | - Lei Yu
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou 510060, China
| | - Hongli Hu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| |
Collapse
|
3
|
Alvin Chew BL, Pan Q, Hu H, Luo D. Structural biology of flavivirus NS1 protein and its antibody complexes. Antiviral Res 2024; 227:105915. [PMID: 38777094 DOI: 10.1016/j.antiviral.2024.105915] [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: 01/30/2024] [Revised: 05/06/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
The genus of flavivirus includes many mosquito-borne human pathogens, such as Zika (ZIKV) and the four serotypes of dengue (DENV1-4) viruses, that affect billions of people as evidenced by epidemics and endemicity in many countries and regions in the world. Among the 10 viral proteins encoded by the viral genome, the nonstructural protein 1 (NS1) is the only secreted protein and has been used as a diagnostic biomarker. NS1 has also been an attractive target for its biotherapeutic potential as a vaccine antigen. This review focuses on the recent advances in the structural landscape of the secreted NS1 (sNS1) and its complex with monoclonal antibodies (mAbs). NS1 forms an obligatory dimer, and upon secretion, it has been reported to be hexametric (trimeric dimers) that could dissociate and bind to the epithelial cell membrane. However, high-resolution structural information has been missing about the high-order oligomeric states of sNS1. Several cryoEM studies have since shown that DENV and ZIKV recombinant sNS1 (rsNS1) are in dynamic equilibrium of dimer-tetramer-hexamer states, with tetramer being the predominant form. It was recently revealed that infection-derived sNS1 (isNS1) forms a complex of the NS1 dimer partially embedded in a High-Density Lipoprotein (HDL) particle. Structures of NS1 in complexes with mAbs have also been reported which shed light on their protective roles during infection. The biological significance of the diversity of NS1 oligomeric states remains to be further studied, to inform future research on flaviviral pathogenesis and the development of therapeutics and vaccines. Given the polymorphism of flavivirus NS1 across sample types with variations in antigenicity, we propose a nomenclature to accurately define NS1 based on the localization and origin.
Collapse
Affiliation(s)
- Bing Liang Alvin Chew
- Lee Kong Chian School of Medicine, Nanyang Technological University, EMB 03-07, 59 Nanyang Drive, Singapore, 636921; NTU Institute of Structural Biology, Nanyang Technological University, EMB 06-01, 59 Nanyang Drive, Singapore, 636921.
| | - Qi Pan
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China.
| | - Hongli Hu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China.
| | - Dahai Luo
- Lee Kong Chian School of Medicine, Nanyang Technological University, EMB 03-07, 59 Nanyang Drive, Singapore, 636921; NTU Institute of Structural Biology, Nanyang Technological University, EMB 06-01, 59 Nanyang Drive, Singapore, 636921; National Centre for Infectious Diseases, Singapore, 308442, Singapore.
| |
Collapse
|
4
|
Chew BLA, Ngoh ANQ, Phoo WW, Chan KWK, Ser Z, Tulsian NK, Lim SS, Weng MJG, Watanabe S, Choy MM, Low J, Ooi EE, Ruedl C, Sobota RM, Vasudevan SG, Luo D. Secreted dengue virus NS1 from infection is predominantly dimeric and in complex with high-density lipoprotein. eLife 2024; 12:RP90762. [PMID: 38787378 PMCID: PMC11126310 DOI: 10.7554/elife.90762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
Severe dengue infections are characterized by endothelial dysfunction shown to be associated with the secreted nonstructural protein 1 (sNS1), making it an attractive vaccine antigen and biotherapeutic target. To uncover the biologically relevant structure of sNS1, we obtained infection-derived sNS1 (isNS1) from dengue virus (DENV)-infected Vero cells through immunoaffinity purification instead of recombinant sNS1 (rsNS1) overexpressed in insect or mammalian cell lines. We found that isNS1 appeared as an approximately 250 kDa complex of NS1 and ApoA1 and further determined the cryoEM structures of isNS1 and its complex with a monoclonal antibody/Fab. Indeed, we found that the major species of isNS1 is a complex of the NS1 dimer partially embedded in a high-density lipoprotein (HDL) particle. Crosslinking mass spectrometry studies confirmed that the isNS1 interacts with the major HDL component ApoA1 through interactions that map to the NS1 wing and hydrophobic domains. Furthermore, our studies demonstrated that the sNS1 in sera from DENV-infected mice and a human patient form a similar complex as isNS1. Our results report the molecular architecture of a biological form of sNS1, which may have implications for the molecular pathogenesis of dengue.
Collapse
Affiliation(s)
- Bing Liang Alvin Chew
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- NTU Institute of Structural Biology, Nanyang Technological UniversitySingaporeSingapore
| | - AN Qi Ngoh
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
| | - Wint Wint Phoo
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and ResearchSingaporeSingapore
| | - Kitti Wing Ki Chan
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
| | - Zheng Ser
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and ResearchSingaporeSingapore
| | - Nikhil K Tulsian
- Department of Biological Sciences, National University of SingaporeSingaporeSingapore
- Singapore Centre for Life Sciences, Department of Biochemistry, National University of SingaporeSingaporeSingapore
| | - Shiao See Lim
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
| | - Mei Jie Grace Weng
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- NTU Institute of Structural Biology, Nanyang Technological UniversitySingaporeSingapore
| | - Satoru Watanabe
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
| | - Milly M Choy
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
| | - Jenny Low
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
- Department of Infectious Diseases, Singapore General HospitalSingaporeSingapore
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
- Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Saw Swee Hock School of Public Health, National University of SingaporeSingaporeSingapore
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological UniversitySingaporeSingapore
| | - Radoslaw M Sobota
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and ResearchSingaporeSingapore
| | - Subhash G Vasudevan
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
- Department of Microbiology and Immunology, National University of SingaporeSingaporeSingapore
- Institute for Glycomics (G26), Griffith University Gold Coast CampusSouthportAustralia
| | - Dahai Luo
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- NTU Institute of Structural Biology, Nanyang Technological UniversitySingaporeSingapore
| |
Collapse
|
5
|
Pan Q, Jiao H, Zhang W, Chen Q, Zhang G, Yu J, Zhao W, Hu H. The step-by-step assembly mechanism of secreted flavivirus NS1 tetramer and hexamer captured at atomic resolution. SCIENCE ADVANCES 2024; 10:eadm8275. [PMID: 38691607 PMCID: PMC11062569 DOI: 10.1126/sciadv.adm8275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 04/01/2024] [Indexed: 05/03/2024]
Abstract
Flaviviruses encode a conserved, membrane-associated nonstructural protein 1 (NS1) with replication and immune evasion functions. The current knowledge of secreted NS1 (sNS1) oligomers is based on several low-resolution structures, thus hindering the development of drugs and vaccines against flaviviruses. Here, we revealed that recombinant sNS1 from flaviviruses exists in a dynamic equilibrium of dimer-tetramer-hexamer states. Two DENV4 hexameric NS1 structures and several tetrameric NS1 structures from multiple flaviviruses were solved at atomic resolution by cryo-EM. The stacking of the tetrameric NS1 and hexameric NS1 is facilitated by the hydrophobic β-roll and connector domains. Additionally, a triacylglycerol molecule located within the central cavity may play a role in stabilizing the hexamer. Based on differentiated interactions between the dimeric NS1, two distinct hexamer models (head-to-head and side-to-side hexamer) and the step-by-step assembly mechanisms of NS1 dimer into hexamer were proposed. We believe that our study sheds light on the understanding of the NS1 oligomerization and contributes to NS1-based therapies.
Collapse
Affiliation(s)
- Qi Pan
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, China
| | - Haizhan Jiao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, China
| | - Wanqin Zhang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, China
| | - Qiang Chen
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, China
| | - Geshu Zhang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, China
| | - Jianhai Yu
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Wei Zhao
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hongli Hu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, China
| |
Collapse
|
6
|
Zeng Q, Liu J, Hao C, Zhang B, Zhang H. Making sense of flavivirus non-strctural protein 1 in innate immune evasion and inducing tissue-specific damage. Virus Res 2023; 336:199222. [PMID: 37716670 PMCID: PMC10518729 DOI: 10.1016/j.virusres.2023.199222] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023]
Abstract
Flaviviruses include medically important mosquito-borne pathogens, such as Zika virus (ZIKV), Japanese encephalitis virus (JEV), dengue virus (DENV) and West Nile virus (WNV), that cause hundreds of millions of infections each year. Currently, there are no approved effect therapies against mosquito-borne flaviviruses. The flaviviruses encoded nonstructural protein 1 (NS1) is a secreted glycoprotein widely involved in viral replication, immune evasion, and directly causing tissue-specific damage during flaviviruses infection. Upon viral infection of host cell, NS1 can be found in multiple oligomeric forms and include a dimer on the cell surface, and a soluble secreted hexameric lipoparticle. In the recent decade, the detailed crystal structure of several flaviviruses NS1 have been determined and unraveled its broader and deeper functions. Consistent with the potential immune function revealed by its structure, NS1 is involved in the escaping of host signal immune pathway mediated by pattern recognition receptors (PRRs), including RIG-I-like receptors (RLRS) and Toll-like receptors (TLRs). Moreover, the flavivirus NS1 is efficiently secreted by infected cells and circulates in the blood of the host to directly induce specific tissues damage. The NS1 of ZIKV, JEV and WNV changes the permeability of brain microvascular endothelial cell to cause endothelial cell dysfunction and promote virus pathogenesis. DENV NS1 can induce systemic tissues damage in humans through multiple strategies. Mutations of several key amino acids in NS1 can reduce the neurovirulence of the flavivirus. In this article, we provide an overview of the latest research on this fascinating protein in these disparate areas.
Collapse
Affiliation(s)
- Quan Zeng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450002, China
| | - Jiaqi Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450002, China
| | - Chenlin Hao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450002, China
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Honglei Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450002, China.
| |
Collapse
|
7
|
Prudencio CR, Gomes da Costa V, Rocha LB, da Costa HHM, Orts DJB, da Silva Santos FR, Rahal P, Lino NAB, da Conceição PJP, Bittar C, Machado RRG, Durigon EL, Araujo JP, Polatto JM, da Silva MA, de Oliveira JA, Mitsunari T, Pereira LR, Andreata-Santos R, de Souza Ferreira LC, Luz D, Piazza RMF. Identification of Zika Virus NS1-Derived Peptides with Potential Applications in Serological Tests. Viruses 2023; 15:v15030654. [PMID: 36992364 PMCID: PMC10052002 DOI: 10.3390/v15030654] [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: 12/21/2022] [Revised: 02/14/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023] Open
Abstract
Zika virus (ZIKV), a mosquito-borne pathogen, is an emerging arbovirus associated with sporadic symptomatic cases of great medical concern, particularly among pregnant women and newborns affected with neurological disorders. Serological diagnosis of ZIKV infection is still an unmet challenge due to the co-circulation of the dengue virus, which shares extensive sequence conservation of structural proteins leading to the generation of cross-reactive antibodies. In this study, we aimed to obtain tools for the development of improved serological tests for the detection of ZIKV infection. Polyclonal sera (pAb) and a monoclonal antibody (mAb 2F2) against a recombinant form of the ZIKV nonstructural protein 1 (NS1) allowed the identification of linear peptide epitopes of the NS1 protein. Based on these findings, six chemically synthesized peptides were tested both in dot blot and ELISA assays using convalescent sera collected from ZIKV-infected patients. Two of these peptides specifically detected the presence of ZIKV antibodies and proved to be candidates for the detection of ZIKV-infected subjects. The availability of these tools opens perspectives for the development of NS1-based serological tests with enhanced sensitivity regarding other flaviviruses.
Collapse
Affiliation(s)
- Carlos Roberto Prudencio
- Laboratório de Imunobiotecnologia, Centro de Imunologia, Instituto Adolfo Lutz, Av. Dr. Arnaldo, 351, São Paulo 01246-902, SP, Brazil
- Correspondence: (C.R.P.); (D.L.); (R.M.F.P.); Tel.: +55-11-3068-2886 (C.R.P.); +55-11-2627-9708 (D.L.); +55-11-2627-9724 (R.M.F.P.)
| | - Vivaldo Gomes da Costa
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Leticia Barboza Rocha
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Hernan Hermes Monteiro da Costa
- Laboratório de Imunobiotecnologia, Centro de Imunologia, Instituto Adolfo Lutz, Av. Dr. Arnaldo, 351, São Paulo 01246-902, SP, Brazil
| | - Diego José Belato Orts
- Laboratório de Imunobiotecnologia, Centro de Imunologia, Instituto Adolfo Lutz, Av. Dr. Arnaldo, 351, São Paulo 01246-902, SP, Brazil
| | - Felipe Rocha da Silva Santos
- Laboratório de Imunobiotecnologia, Centro de Imunologia, Instituto Adolfo Lutz, Av. Dr. Arnaldo, 351, São Paulo 01246-902, SP, Brazil
| | - Paula Rahal
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Nikolas Alexander Borsato Lino
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Pâmela Jóyce Previdelli da Conceição
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Cintia Bittar
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Rafael Rahal Guaragna Machado
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Edison Luiz Durigon
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - João Pessoa Araujo
- Instituto de Biotecnologia, Universidade Estadual Paulista Júlio de Mesquita Filho, Botucatu 18607-440, SP, Brazil
| | - Juliana Moutinho Polatto
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Miriam Aparecida da Silva
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Joyce Araújo de Oliveira
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Thais Mitsunari
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Lennon Ramos Pereira
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Robert Andreata-Santos
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Luís Carlos de Souza Ferreira
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
- Plataforma Científica Pasteur USP, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Daniela Luz
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
- Correspondence: (C.R.P.); (D.L.); (R.M.F.P.); Tel.: +55-11-3068-2886 (C.R.P.); +55-11-2627-9708 (D.L.); +55-11-2627-9724 (R.M.F.P.)
| | - Roxane Maria Fontes Piazza
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
- Correspondence: (C.R.P.); (D.L.); (R.M.F.P.); Tel.: +55-11-3068-2886 (C.R.P.); +55-11-2627-9708 (D.L.); +55-11-2627-9724 (R.M.F.P.)
| |
Collapse
|
8
|
Molecular Mechanism and Role of Japanese Encephalitis Virus Infection in Central Nervous System-Mediated Diseases. Viruses 2022; 14:v14122686. [PMID: 36560690 PMCID: PMC9781168 DOI: 10.3390/v14122686] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
The Japanese encephalitis virus (JEV) is the most common cause of neurodegenerative disease in Southeast Asia and the Western Pacific region; approximately 1.15 billion people are at risk, and thousands suffer from permanent neurological disorders across Asian countries, with 10-15 thousand people dying each year. JEV crosses the blood-brain barrier (BBB) and forms a complex with receptors on the surface of neurons. GRP78, Src, TLR7, caveolin-1, and dopamine receptor D2 are involved in JEV binding and entry into the neurons, and these receptors also play a role in carcinogenic activity in cells. JEV binds to GRP78, a member of the HSP70 overexpressed on malignant cells to enter neurons, indicating a higher chance of JEV infection in cancer patients. However, JEV enters human brain microvascular endothelial cells via an endocytic pathway mediated by caveolae and the ezrin protein and also targets dopamine-rich areas for infection of the midbrain via altering dopamine levels. In addition, JEV complexed with CLEC5A receptor of macrophage cells is involved in the breakdown of the BBB and central nervous system (CNS) inflammation. CLEC5A-mediated infection is also responsible for the influx of cytokines into the CNS. In this review, we discuss the neuronal and macrophage surface receptors involved in neuronal death.
Collapse
|
9
|
Roberts A, Kesarwani V, Gupta R, Gandhi S. Electroactive reduced graphene oxide for highly sensitive detection of secretory non-structural 1 protein: A potential diagnostic biomarker for Japanese encephalitis virus. Biosens Bioelectron 2022; 198:113837. [PMID: 34864242 DOI: 10.1016/j.bios.2021.113837] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/11/2021] [Accepted: 11/24/2021] [Indexed: 12/13/2022]
Abstract
Fluorine Doped Tin Oxide (FTO) electrode was fabricated with reduced Graphene Oxide (rGO) for sensitive detection of Japanese encephalitis virus (JEV) non-structural 1 (NS1) protein. Beforehand, in-silico 3D structure, stability, and docking of recombinant JEV NS1 antigen (NS1-Ag) and antibody (Ab) was evaluated. The recombinant NS1 Ag of 42 kDa was produced in-house by successful cloning into pET-28a(+) plasmid and further expressed using BL21 Escherichia coli (E. coli) cells. The NS1 Ag was used to raise polyclonal antibodies (Ab) and both were characterized via Sodium Dodecyl Sulphate Polyacrylamide Gel Electrophoresis (SDS-PAGE), Western Blot, Matrix-Assisted Laser Desorption/Ionization-Time of Flight (MALDI-TOF), and Enzyme-Linked Immunosorbent Assay (ELISA). Further characterisation of all binding events such as rGO synthesis, and its conjugation with NS1 Ab, and NS1 Ag were confirmed through Fourier-Transform Infrared Spectroscopy (FTIR), Raman Spectroscopy, Energy Dispersive X-Ray Analysis (EDX), Scanning Electron Microscopy (SEM), Cyclic Voltammetry (CV) and Differential Pulse Voltammetry (DPV). The fabricated FTO electrode was optimised for various parameters such as pH, response time, temperature, concentration, and scan rate. The detection of JEV NS1 Ag was performed in buffer (LOD- 0.92 fM) as well in spiked serum (LOD- 1.3 fM) samples. The JEV NS1 Ab showed negligible cross-reactivity with other flaviviral NS1 Ag, provided a rapid response within 5 s, and remained stable up to 4 weeks. Furthermore, the fabricated immunosensor may be a potential candidate for further miniaturisation for accurate and early diagnosis of JEV in clinical samples.
Collapse
Affiliation(s)
- Akanksha Roberts
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad, 500032, Telangana, India; DBT-Regional Centre for Biotechnology (RCB), Faridabad, 121001, NCR Delhi, India
| | - Veerbhan Kesarwani
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad, 500032, Telangana, India
| | - Rupal Gupta
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad, 500032, Telangana, India
| | - Sonu Gandhi
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad, 500032, Telangana, India; DBT-Regional Centre for Biotechnology (RCB), Faridabad, 121001, NCR Delhi, India.
| |
Collapse
|
10
|
Sharma KB, Vrati S, Kalia M. Pathobiology of Japanese encephalitis virus infection. Mol Aspects Med 2021; 81:100994. [PMID: 34274157 DOI: 10.1016/j.mam.2021.100994] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 12/25/2022]
Abstract
Japanese encephalitis virus (JEV) is a flavivirus, spread by the bite of carrier Culex mosquitoes. The subsequent disease caused is Japanese encephalitis (JE), which is the leading global cause of virus-induced encephalitis. The disease is predominant in the entire Asia-Pacific region with the potential of global spread. JEV is highly neuroinvasive with symptoms ranging from mild fever to severe encephalitis and death. One-third of JE infections are fatal, and half of the survivors develop permanent neurological sequelae. Disease prognosis is determined by a series of complex and intertwined signaling events dictated both by the virus and the host. All flaviviruses, including JEV replicate in close association with ER derived membranes by channelizing the protein and lipid components of the ER. This leads to activation of acute stress responses in the infected cell-oxidative stress, ER stress, and autophagy. The host innate immune and inflammatory responses also enter the fray, the components of which are inextricably linked to the cellular stress responses. These are especially crucial in the periphery for dendritic cell maturation and establishment of adaptive immunity. The pathogenesis of JEV is a combination of direct virus induced neuronal cell death and an uncontrolled neuroinflammatory response. Here we provide a comprehensive review of the JEV life cycle and how the cellular stress responses dictate the pathobiology and resulting immune response. We also deliberate on how modulation of these stress pathways could be a potential strategy to develop therapeutic interventions, and define the persisting challenges.
Collapse
Affiliation(s)
- Kiran Bala Sharma
- Virology Research Group, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India
| | - Sudhanshu Vrati
- Virology Research Group, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India.
| | - Manjula Kalia
- Virology Research Group, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India.
| |
Collapse
|
11
|
Wang P, Liu X, Li Q, Wang J, Ruan W. Proteomic analyses identify intracellular targets for Japanese encephalitis virus nonstructural protein 1 (NS1). Virus Res 2021; 302:198495. [PMID: 34175344 DOI: 10.1016/j.virusres.2021.198495] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 11/25/2022]
Abstract
Japanese encephalitis is a zoonotic disease caused by Japanese encephalitis virus (JEV). JEV nonstructural protein 1 (NS1) is involved in many crucial biological events during viral infection and immune suppression. To investigate the role of JEV NS1 in virus-infected cells, the molecules with which it interacts intracellularly were screened with a pull-down assay and liquid chromatography-tandem mass spectrometry (LC-MS/MS). The interaction between heterogeneous nuclear ribonucleoprotein K (hnRNP K), vimentin and NS1 were verified with coimmunoprecipitation and confocal assays. Our results show that JEV NS1 interacts with vimentin, hnRNP K and colocalizes with cellular vimentin and hnRNP K. Furthermore, our results demonstrate that the expression of vimentin and hnRNP K were up-regulated in both NS1-transfected and JEV-infected cells. Knocking down vimentin and hnRNP K reduced viral replication while conversely, over-expression of vimentin and hnRNP K improved viral replication, suggesting an important role for this protein in the viral life cycle. Also, We found that vimentin also interacted with hnRNP K after overexpression of NS1 or JEV infection. These findings provide insight into the molecular mechanism of JEV replication and highlight the key role the NS1 in JEV infection.
Collapse
Affiliation(s)
- Peng Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Xinze Liu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Qi Li
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Jue Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Wenke Ruan
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China.
| |
Collapse
|
12
|
Carpio KL, Barrett ADT. Flavivirus NS1 and Its Potential in Vaccine Development. Vaccines (Basel) 2021; 9:622. [PMID: 34207516 PMCID: PMC8229460 DOI: 10.3390/vaccines9060622] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/19/2022] Open
Abstract
The Flavivirus genus contains many important human pathogens, including dengue, Japanese encephalitis (JE), tick-borne encephalitis (TBE), West Nile (WN), yellow fever (YF) and Zika (ZIK) viruses. While there are effective vaccines for a few flavivirus diseases (JE, TBE and YF), the majority do not have vaccines, including WN and ZIK. The flavivirus nonstructural 1 (NS1) protein has an unusual structure-function because it is glycosylated and forms different structures to facilitate different roles intracellularly and extracellularly, including roles in the replication complex, assisting in virus assembly, and complement antagonism. It also plays a role in protective immunity through antibody-mediated cellular cytotoxicity, and anti-NS1 antibodies elicit passive protection in animal models against a virus challenge. Historically, NS1 has been used as a diagnostic marker for the flavivirus infection due to its complement fixing properties and specificity. Its role in disease pathogenesis, and the strong humoral immune response resulting from infection, makes NS1 an excellent target for inclusion in candidate flavivirus vaccines.
Collapse
Affiliation(s)
- Kassandra L. Carpio
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Alan D. T. Barrett
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
13
|
Molecular Insights into the Flavivirus Replication Complex. Viruses 2021; 13:v13060956. [PMID: 34064113 PMCID: PMC8224304 DOI: 10.3390/v13060956] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
Flaviviruses are vector-borne RNA viruses, many of which are clinically relevant human viral pathogens, such as dengue, Zika, Japanese encephalitis, West Nile and yellow fever viruses. Millions of people are infected with these viruses around the world each year. Vaccines are only available for some members of this large virus family, and there are no effective antiviral drugs to treat flavivirus infections. The unmet need for vaccines and therapies against these flaviviral infections drives research towards a better understanding of the epidemiology, biology and immunology of flaviviruses. In this review, we discuss the basic biology of the flavivirus replication process and focus on the molecular aspects of viral genome replication. Within the virus-induced intracellular membranous compartments, flaviviral RNA genome replication takes place, starting from viral poly protein expression and processing to the assembly of the virus RNA replication complex, followed by the delivery of the progeny viral RNA to the viral particle assembly sites. We attempt to update the latest understanding of the key molecular events during this process and highlight knowledge gaps for future studies.
Collapse
|
14
|
Wan J, Wang T, Xu J, Ouyang T, Wang Q, Zhang Y, Weng S, Li Y, Wang Y, Xin X, Wang X, Li S, Kong L. Novel Japanese encephalitis virus NS1-based vaccine: Truncated NS1 fused with E. coli heat labile enterotoxin B subunit. EBioMedicine 2021; 67:103353. [PMID: 33971403 PMCID: PMC8122160 DOI: 10.1016/j.ebiom.2021.103353] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/06/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Current vaccines against Japanese encephalitis virus (JEV) of flaviviruses have some disadvantages, such as the risk of virulent reversion. Non-structural protein NS1 is conserved among flaviviruses and confers immune protection without the risk of antibody-dependent enhancement (ADE). Therefore, NS1 has become a promising vaccine candidate against flaviviruses. METHODS A NS1-based vaccine (LTB-NS1∆63) with a truncated NS1 protein (NS1∆63) fused to E. coli heat-labile enterotoxin B subunit (LTB) was expressed in E.coli and explored for its ability to induce immune responses. Safety of LTB-NS1∆63 was assessed by determining its toxicity in vitro and in vivo. Protective capability of LTB-NS1∆63 and its-induced antisera was evaluated in the mice challenged with JEV by analyzing mortality and morbidity. FINDINGS LTB-NS1∆63 induced immune responses to a similar level as LTB-NS1, but more robust than NS1∆63 alone, particularly in the context of oral immunization of mice. Oral vaccination of LTB-NS1∆63 led to a higher survival rate than that of NS1∆63 or live-attenuated JEV vaccine SA14-14-2 in the mice receiving lethal JEV challenge. LTB-NS1∆63 protein also significantly decreases the morbidity of JEV-infected mice. In addition, passive transfer of LTB-NS1∆63-induced antisera provides a protection against JEV infection in mice. INTERPRETATION NS1∆63 bears JEV NS1 antigenicity. Besides, LTB-NS1∆63 could serve as a novel protein-based mucosa vaccine targeting JEV and other flaviviruses. FUNDING This work was supported by the National Natural Science Foundation, Jiangxi Province Science and Technology Committee, Education Department of Jiangxi Province.
Collapse
Affiliation(s)
- Jiawu Wan
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Ting Wang
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Jing Xu
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Tao Ouyang
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Qianruo Wang
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Yanni Zhang
- Jiangxi Province Center for Disease Control and Prevention, Nanchang, Jiangxi, China
| | - Shiqi Weng
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Yihan Li
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Yu Wang
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Xiu Xin
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Xiaoling Wang
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Sha Li
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China.
| | - Lingbao Kong
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China.
| |
Collapse
|
15
|
Chavas LMG, Gourhant P, Guimaraes BG, Isabet T, Legrand P, Lener R, Montaville P, Sirigu S, Thompson A. PROXIMA-1 beamline for macromolecular crystallography measurements at Synchrotron SOLEIL. JOURNAL OF SYNCHROTRON RADIATION 2021; 28:970-976. [PMID: 33950005 PMCID: PMC8127379 DOI: 10.1107/s1600577521002605] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 03/09/2021] [Indexed: 05/07/2023]
Abstract
The undulator beamline PROXIMA-1 at Synchrotron SOLEIL scheduled its first users in March 2008. The endstation is dedicated to biomolecular crystallography experiments, with a layout designed to favour anomalous data recording and studies of crystals with large cell dimensions. In 12 years, the beamline has accommodated 4267 shifts of 8 h and more than 6300 visitors. By the end of 2020, it saw 1039 identified published scientific papers referring to 1415 coordinates deposited in the Protein Data Bank. The current paper describes the PROXIMA-1 beamline, including the recent specific implementations developed for the sample environment. The setup installed in the experimental station contains numerous beam-shaping equipment, a chi-geometry three-axis goniometer, a single-photon-counting pixel-array X-ray detector, combined with a medium-throughput sample exchange robot. As part of a standard experimental scheme, PROXIMA-1 can also be accessed via `mail-in' services or remotely.
Collapse
Affiliation(s)
- Leonard M. G. Chavas
- Synchrotron SOLEIL, 91192 Gif-sur-Yvette, France
- Nagoya University, Nagoya 464-8603, Japan
| | | | | | | | | | - Robin Lener
- Synchrotron SOLEIL, 91192 Gif-sur-Yvette, France
| | | | | | | |
Collapse
|
16
|
Modhiran N, Song H, Liu L, Bletchly C, Brillault L, Amarilla AA, Xu X, Qi J, Chai Y, Cheung STM, Traves R, Setoh YX, Bibby S, Scott CAP, Freney ME, Newton ND, Khromykh AA, Chappell KJ, Muller DA, Stacey KJ, Landsberg MJ, Shi Y, Gao GF, Young PR, Watterson D. A broadly protective antibody that targets the flavivirus NS1 protein. Science 2021; 371:190-194. [PMID: 33414219 DOI: 10.1126/science.abb9425] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022]
Abstract
There are no approved flaviviral therapies and the development of vaccines against flaviruses has the potential of being undermined by antibody-dependent enhancement (ADE). The flavivirus nonstructural protein 1 (NS1) is a promising vaccine antigen with low ADE risk but has yet to be explored as a broad-spectrum therapeutic antibody target. Here, we provide the structural basis of NS1 antibody cross-reactivity through cocrystallization of the antibody 1G5.3 with NS1 proteins from dengue and Zika viruses. The 1G5.3 antibody blocks multi-flavivirus NS1-mediated cell permeability in disease-relevant cell lines, and therapeutic application of 1G5.3 reduces viremia and improves survival in dengue, Zika, and West Nile virus murine models. Finally, we demonstrate that 1G5.3 protection is independent of effector function, identifying the 1G5.3 epitope as a key site for broad-spectrum antiviral development.
Collapse
Affiliation(s)
- Naphak Modhiran
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Hao Song
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Lidong Liu
- Division of Laboratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cheryl Bletchly
- Microbiology, Pathology Queensland, Queensland Health, Herston, Queensland, Australia
| | - Lou Brillault
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia.,Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Australia
| | - Alberto A Amarilla
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Xiaoying Xu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Yan Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Stacey T M Cheung
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Renee Traves
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Yin Xiang Setoh
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Summa Bibby
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Connor A P Scott
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Morgan E Freney
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Natalee D Newton
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Alexander A Khromykh
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Keith J Chappell
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - David A Muller
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Katryn J Stacey
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Michael J Landsberg
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Yi Shi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| | - George F Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China. .,Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Paul R Young
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia.
| | - Daniel Watterson
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
17
|
Biering SB, Akey DL, Wong MP, Brown WC, Lo NTN, Puerta-Guardo H, Tramontini Gomes de Sousa F, Wang C, Konwerski JR, Espinosa DA, Bockhaus NJ, Glasner DR, Li J, Blanc SF, Juan EY, Elledge SJ, Mina MJ, Beatty PR, Smith JL, Harris E. Structural basis for antibody inhibition of flavivirus NS1-triggered endothelial dysfunction. Science 2021; 371:194-200. [PMID: 33414220 PMCID: PMC8000976 DOI: 10.1126/science.abc0476] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 11/23/2020] [Indexed: 01/01/2023]
Abstract
Medically important flaviviruses cause diverse disease pathologies and collectively are responsible for a major global disease burden. A contributing factor to pathogenesis is secreted flavivirus nonstructural protein 1 (NS1). Despite demonstrated protection by NS1-specific antibodies against lethal flavivirus challenge, the structural and mechanistic basis remains unknown. Here, we present three crystal structures of full-length dengue virus NS1 complexed with a flavivirus-cross-reactive, NS1-specific monoclonal antibody, 2B7, at resolutions between 2.89 and 3.96 angstroms. These structures reveal a protective mechanism by which two domains of NS1 are antagonized simultaneously. The NS1 wing domain mediates cell binding, whereas the β-ladder triggers downstream events, both of which are required for dengue, Zika, and West Nile virus NS1-mediated endothelial dysfunction. These observations provide a mechanistic explanation for 2B7 protection against NS1-induced pathology and demonstrate the potential of one antibody to treat infections by multiple flaviviruses.
Collapse
Affiliation(s)
- Scott B Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - David L Akey
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marcus P Wong
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - W Clay Brown
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nicholas T N Lo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Henry Puerta-Guardo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | | | - Chunling Wang
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Jamie R Konwerski
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Diego A Espinosa
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Nicholas J Bockhaus
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Dustin R Glasner
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Jeffrey Li
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Sophie F Blanc
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Evan Y Juan
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Stephen J Elledge
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, and Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Michael J Mina
- Center for Communicable Disease Dynamics, Department of Epidemiology, and Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA 02115, USA
| | - P Robert Beatty
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Janet L Smith
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA.
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA.
| |
Collapse
|
18
|
Bhardwaj T, Saumya KU, Kumar P, Sharma N, Gadhave K, Uversky VN, Giri R. Japanese encephalitis virus - exploring the dark proteome and disorder-function paradigm. FEBS J 2020; 287:3751-3776. [PMID: 32473054 DOI: 10.1111/febs.15427] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/26/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022]
Abstract
Japanese encephalitis virus (JEV) is one of the major causes of viral encephalitis all around the globe. Approximately 3 billion people in endemic areas are at risk of Japanese encephalitis. To develop a wholistic understanding of the viral proteome, it is important to investigate both its ordered and disordered proteins. However, the functional and structural significance of disordered regions in the JEV proteome has not been systematically investigated as of yet. To fill this gap, we used here a set of bioinformatics tools to analyze the JEV proteome for the predisposition of its proteins for intrinsic disorder and for the presence of the disorder-based binding regions (also known as molecular recognition features, MoRFs). We also analyzed all JEV proteins for the presence of the probable nucleic acid-binding (DNA and RNA) sites. The results of these computational studies are experimentally validated using JEV capsid protein as an illustrative example. In agreement with bioinformatic analysis, we found that the N-terminal region of the JEV capsid (residues 1-30) is intrinsically disordered. We showed that this region is characterized by the temperature response typical for highly disordered proteins. Furthermore, we have experimentally shown that this disordered N-terminal domain of a capsid protein has a noticeable 'gain-of-structure' potential. In addition, using DOPS liposomes, we demonstrated the presence of pronounced membrane-mediated conformational changes in the N-terminal region of JEV capsid. In our view, this disorder-centric analysis would be helpful for a better understanding of the JEV pathogenesis.
Collapse
Affiliation(s)
- Taniya Bhardwaj
- School of Basic Sciences, Indian Institute of Technology Mandi, India
| | - Kumar Udit Saumya
- School of Basic Sciences, Indian Institute of Technology Mandi, India
| | - Prateek Kumar
- School of Basic Sciences, Indian Institute of Technology Mandi, India
| | - Nitin Sharma
- School of Basic Sciences, Indian Institute of Technology Mandi, India
| | - Kundlik Gadhave
- School of Basic Sciences, Indian Institute of Technology Mandi, India
| | - Vladimir N Uversky
- Department of Molecular Medicine, USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Laboratory of New Methods in Biology, Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center 'Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences', Russia
| | - Rajanish Giri
- School of Basic Sciences, Indian Institute of Technology Mandi, India
| |
Collapse
|
19
|
Anwar MN, Wang X, Hameed M, Wahaab A, Li C, Sharma M, Pang L, Malik MI, Liu K, Li B, Qiu Y, Wei J, Ma Z. Phenotypic and Genotypic Comparison of a Live-Attenuated Genotype I Japanese Encephalitis Virus SD12-F120 Strain with Its Virulent Parental SD12 Strain. Viruses 2020; 12:v12050552. [PMID: 32429445 PMCID: PMC7290960 DOI: 10.3390/v12050552] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/05/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
The phenotypic and genotypic characteristics of a live-attenuated genotype I (GI) strain (SD12-F120) of Japanese encephalitis virus (JEV) were compared with its virulent parental SD12 strain to gain an insight into the genetic changes acquired during the attenuation process. SD12-F120 formed smaller plaque on BHK-21 cells and showed reduced replication in mouse brains compared with SD12. Mice inoculated with SD12-F120 via either intraperitoneal or intracerebral route showed no clinical symptoms, indicating a highly attenuated phenotype in terms of both neuroinvasiveness and neurovirulence. SD12-F120 harbored 29 nucleotide variations compared with SD12, of which 20 were considered silent nucleotide mutations, while nine resulted in eight amino acid substitutions. Comparison of the amino acid variations of SD12-F120 vs. SD12 pair with those from other four isogenic pairs of the attenuated and their virulent parental strains revealed that the variations at E138 and E176 positions of E protein were identified in four and three pairs, respectively, while the remaining amino acid variations were almost unique to their respective strain pairs. These observations suggest that the genetic changes acquired during the attenuation process were likely to be strain-specific and that the mechanisms associated with JEV attenuation/virulence are complicated.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jianchao Wei
- Correspondence: (J.W.); (Z.M.); Tel.: +86-21-3468-3635 (J.W.); +86-21-34293139 (Z.M.); Fax: +86-21-54081818 (J.W. & Z.M.)
| | - Zhiyong Ma
- Correspondence: (J.W.); (Z.M.); Tel.: +86-21-3468-3635 (J.W.); +86-21-34293139 (Z.M.); Fax: +86-21-54081818 (J.W. & Z.M.)
| |
Collapse
|
20
|
Abstract
Japanese encephalitis (JE) is a clinical manifestation of the brain inflammation caused by JE virus (JEV). This virus imparts permanent neurological damage, thus imposing a heavy burden on public health and society. Neuro-inflammation is the hallmark of JEV infection. The prolonged pro-inflammatory response is due primarily to microglial activation, which eventually leads to severe encephalitis. A continual effort is going on in the scientific community toward an understanding of cellular and molecular factors that are involved in JEV neuro-invasion and inflammatory processes. This review not only gives a comprehensive update on the recent advances on understanding virus structure and mechanisms of pathogenesis but also briefly discusses crucial unresolved issues. We also highlight challenging areas of research that might open new avenues for controlling virus-induced neuro-inflammation.
Collapse
Affiliation(s)
- Arup Banerjee
- Laboratory of Virology, Regional Centre for Biotechnology, Faridabad, Haryana, India.,Translational Health Science & Technology Institute, Faridabad, Haryana, India
| | - Aarti Tripathi
- Translational Health Science & Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
21
|
Reyes-Sandoval A, Ludert JE. The Dual Role of the Antibody Response Against the Flavivirus Non-structural Protein 1 (NS1) in Protection and Immuno-Pathogenesis. Front Immunol 2019; 10:1651. [PMID: 31379848 PMCID: PMC6657369 DOI: 10.3389/fimmu.2019.01651] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/03/2019] [Indexed: 12/22/2022] Open
Abstract
Dengue and Zika viruses are closely related mosquito-borne flaviviruses responsible for major public health problems in tropical and sub-tropical countries. The genomes of both, dengue and zika viruses encodes 10 genes that are translated into three structural proteins (C, prM, and E) and seven non-structural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5). The non-structural protein 1 (NS1) is a highly conserved glycoprotein of approximately 48–50 KDa. In infected cells, NS1 is found as a homodimer associated with intracellular membranes and replication complexes, serving as a scaffolding protein in virus replication and morphogenesis. NS1 is secreted efficiently from infected cells as a hexamer and is found in patient's sera during the acute phase of the disease. NS1 detection in sera is a valuable diagnostic marker and immunization with NS1 has been shown to protect animal models from lethal challenges with dengue and Zika viruses. Nevertheless, soluble NS1 has been associated with severe dengue and anti-NS1 antibodies have been reported to cross-react with host platelets and endothelial cells and thus presumably contribute to pathogenesis. Due to the implications of NS1 in arbovirus pathogenesis and its relevance as vaccine candidate, we discuss the dual role that anti-NS1 antibodies may play in protection and disease and the challenges that need to be overcome to develop safe and effective NS1-based vaccines against dengue and Zika.
Collapse
Affiliation(s)
- Arturo Reyes-Sandoval
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Juan E Ludert
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico
| |
Collapse
|
22
|
Poonsiri T, Wright GSA, Solomon T, Antonyuk SV. Crystal Structure of the Japanese Encephalitis Virus Capsid Protein. Viruses 2019; 11:E623. [PMID: 31284608 PMCID: PMC6669762 DOI: 10.3390/v11070623] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/01/2019] [Accepted: 07/04/2019] [Indexed: 12/29/2022] Open
Abstract
Japanese encephalitis (JE) is inflammation and swelling of the brain caused by the JE virus (JEV), a mosquito-borne member of the Flavivirus family. There are around 68,000 JE cases worldwide each year, many of which result in permanent brain damage and death. There is no specific treatment for JE. Here we present the crystal structure of the JEV capsid protein, a potential drug target, at 1.98 Å, and compare it to other flavivirus capsid proteins. The JEV capsid has a helical secondary structure (α helixes 1-4) and a similar protein fold to the dengue virus (DENV), the West Nile virus (WNV), and the Zika virus (ZIKV) capsid proteins. It forms a homodimer by antiparallel pairing with another subunit (') through α-helix 1-1', 2-2', and 4-4' interactions. This dimeric form is believed to be the building block of the nucleocapsid. The flexibility of the N-terminal α helix-1 allows the formation of closed and open conformations with possible functional importance. The basic C-terminal pairing of α4-4' forms a coiled-coil-like structure, indicating possible nucleic acid binding functionality. However, a comparison with other nucleic acid interacting domains indicates that homodimerization would preclude binding. This is the first JEV capsid protein to be described and is an addition to the structural biology of the Flavivirus.
Collapse
Affiliation(s)
- Thanalai Poonsiri
- Molecular Biophysics Group, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, L69 7ZB Liverpool, UK
- Health Protection Research Unit on Emerging and Zoonotic Infections, Institute of Infection and Global Health, University of Liverpool, L69 7BE Liverpool, UK
| | - Gareth S A Wright
- Molecular Biophysics Group, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, L69 7ZB Liverpool, UK
| | - Tom Solomon
- Health Protection Research Unit on Emerging and Zoonotic Infections, Institute of Infection and Global Health, University of Liverpool, L69 7BE Liverpool, UK
- Walton Centre NHS Foundation Trust, L9 7LJ Liverpool, UK
| | - Svetlana V Antonyuk
- Molecular Biophysics Group, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, L69 7ZB Liverpool, UK.
| |
Collapse
|
23
|
Dual targeting of dengue virus virions and NS1 protein with the heparan sulfate mimic PG545. Antiviral Res 2019; 168:121-127. [PMID: 31085206 DOI: 10.1016/j.antiviral.2019.05.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/16/2019] [Accepted: 05/09/2019] [Indexed: 01/08/2023]
Abstract
Dengue virus (DENV) is the most prevalent mosquito-borne flavivirus that infects humans. At present, there are no specific antiviral drugs to treat DENV infection and vaccine development has met with challenges. DENV encodes two glycosaminoglycan (GAG) binding proteins; Envelope (E) and non-structural protein 1 (NS1). While previous work has validated the use of GAG analogues as inhibitors of E mediated virus-cell attachment, their potential for antiviral intervention in NS1 protein toxicity has not yet been explored. Here, we investigate the potential of the heparan sulfate mimetic PG545 as a dual purpose compound to target both DENV virion infectivity and NS1 function. In comparison to a non-sulfated analogue, we show that PG545 potently inhibits DENV infectivity with no cytotoxic effect. Against NS1, PG545 completely blocks the induction of cellular activation and abolishes NS1-mediated disruption of endothelial monolayer integrity. Furthermore, PG545 treatment moderately improves survival from lethal DENV challenge in a murine model. At peak disease, PG545-treated mice have lower viremia, circulating NS1 and serum TNF-α. Consistent with anti-NS1 activity, PG545 treatment also reduces systemic vascular leakage caused by DENV infection in vivo. Taken together, these findings demonstrate that the dual targeting of DENV virions and NS1 using GAG analogues offers a new avenue for DENV drug development.
Collapse
|
24
|
Differential replication efficiencies between Japanese encephalitis virus genotype I and III in avian cultured cells and young domestic ducklings. PLoS Negl Trop Dis 2018; 12:e0007046. [PMID: 30562354 PMCID: PMC6314627 DOI: 10.1371/journal.pntd.0007046] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/02/2019] [Accepted: 12/02/2018] [Indexed: 12/28/2022] Open
Abstract
Japanese encephalitis virus (JEV) genotype dominance has shifted to genotype I (GI) from genotype III (GIII) in China as demonstrated by molecular epidemiological surveillance. In this study, we performed a serological survey in JEV-non-vaccinated pigs to confirm JEV genotype shift at the sero-epidemiological level. The average ratio of GI/GIII infection was 1.87, suggesting co-circulation of GI and GIII infections with GI infection being more prevalent in pigs in China. To gain an insight into the reasons for this JEV genotype shift, the replication kinetics of seven recently-isolated JEV isolates including three GI strains and four GIII strains were compared in mosquito C6/36 cells, chicken fibroblast cells (DF-1) and porcine iliac artery endothelial cells (PIEC). We observed that GI strains replicated more efficiently than GIII strains in DF-1 and PIEC cells, particularly in DF-1 cells with titers reaching 22.9–225.3 fold higher than GIII strains. This shows an enhanced replication efficiency of GI viruses in avian cells. To examine this enhanced replication efficiency in vivo, young domestic ducklings were used as the animal model and inoculated with GI and GIII strains at day 2 post-hatching. We observed that GI-inoculated ducklings developed higher viremia titers and displayed a comparatively longer viremic duration than GIII-inoculated ducklings. These results conform to the hypothesis of an enhanced replication efficiency for GI viruses in birds. There are 36 amino acid differences between GI and GIII viruses, some of which may be responsible for the enhanced replication efficiency of GI viruses in birds. Based on these findings, we speculated that the enhanced replication of GI viruses in birds would have resulted in higher exposure and therefore infection in mosquitoes, which could result in an increased transmission efficiency of GI viruses in the birds-mosquitoes-birds enzootic transmission cycle, thereby contributing to JEV genotype shift. Japanese encephalitis virus (JEV) causes encephalitis in humans and reproductive disorder in pigs. The enzootic transmission cycle of JEV is maintained in nature by several species of mosquitoes and vertebrates including birds and pigs. In recent years, JEV genotype I (GI) replaced genotype IIII (GIII) as the dominant genotype in Asian countries. Genotype shift has an impact on disease control, and understanding the reasons for this shift will offer valuable insight into avenues for future disease control. Therefore, we compared the replication efficiencies of GI and GIII viruses in vitro and in vivo. We observed that GI viruses show higher replication titers in avian cells and higher viremia levels in young domestic ducklings than GIII viruses, suggesting an enhanced replication efficiency of GI viruses in birds. Based on these findings, we speculated that the enhanced replication of GI viruses in birds could provide increased mosquito infection, leading to an increase in the birds-mosquitoes-birds transmission cycle, thereby contributing to JEV genotype shift.
Collapse
|