1
|
Ugwu CC, Hair-Bejo M, Nurulfiza MI, Omar AR, Ideris A. Attenuation and molecular characterization of fowl adenovirus 8b propagated in a bioreactor and its immunogenicity, efficacy, and virus shedding in broiler chickens. Vet World 2024; 17:744-755. [PMID: 38798289 PMCID: PMC11111708 DOI: 10.14202/vetworld.2024.744-755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/05/2024] [Indexed: 05/29/2024] Open
Abstract
Background and Aim Live-attenuated vaccines are the most successful type of vaccine and could be useful in controlling fowl adenovirus (FAdV) 8b infection. This study aimed to attenuate, molecularly characterize, and determine the immunogenicity, efficacy, and challenge virus shedding in broiler chickens. Materials and Methods The FAdV 8b isolate (UPM08136) was passaged onto chicken embryo liver (CEL) cells until attenuation. We sequenced and analyzed the hexon and fiber genes of the passage isolates. The attenuated bioreactor-passage isolate was inoculated into 1-day-old broiler chickens with (attenuated and inactivated) and without booster groups and challenged. Body weight (BW), liver weight (LW), liver: body weight ratio (LBR), FAdV antibody titers, T-lymphocyte subpopulation in the liver, spleen, and thymus, and challenge virus load and shedding were measured. Results Typical cytopathic effects with novel genetic changes on CEL cells were observed. The uninoculated control-challenged (UCC) group had significantly lower BW and higher LW and LBR than the inoculated groups. A significantly higher FAdV antibody titer was observed in the challenged non-booster and attenuated booster groups than in the UCC group. T cells in the spleen and thymus of the liver of inoculated chickens were higher than uninoculated control group levels at all-time points and at different times. A significantly higher FAdV challenge virus load was observed in the liver and shedding in the cloaca of UCC chickens than in non-booster chickens. Conclusion The FAdV 8b isolate was successfully attenuated, safe, and immunogenic. It reduces virus shedding and is effective and recommended as a vaccine against FAdV infection in broiler chickens.
Collapse
Affiliation(s)
- Chidozie C. Ugwu
- Department of Veterinary Pathology and Microbiology Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Department of Animal Science and Technology, Federal University of Technology, Owerri 460114, Imo State, Nigeria
| | - Mohd Hair-Bejo
- Department of Veterinary Pathology and Microbiology Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Laboratory of Vaccine and Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Mat I. Nurulfiza
- Laboratory of Vaccine and Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Abdul R. Omar
- Department of Veterinary Pathology and Microbiology Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Laboratory of Vaccine and Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Aini Ideris
- Department of Veterinary Pathology and Microbiology Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Laboratory of Vaccine and Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
2
|
Tan N, Chen C, Ren Y, Huang R, Zhu Z, Xu K, Yang X, Yang J, Yuan L. Nucleotide at position 66 of NS2A in Japanese encephalitis virus is associated with the virulence and proliferation of virus. Virus Genes 2024; 60:9-17. [PMID: 37938470 DOI: 10.1007/s11262-023-02036-5] [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: 08/09/2023] [Accepted: 10/10/2023] [Indexed: 11/09/2023]
Abstract
Most wild strains of Japanese encephalitis virus (JEV) produce NS1' protein, which plays an important role in viral infection and immune escape. The G66A nucleotide mutation in NS2A gene of the wild strain SA14 prevented the ribosomal frameshift that prevented the production of NS1' protein, thus reduced the virulence. In this study, the 66th nucleotide of the NS2A gene of SA14 was mutated into A, U or C, respectively. Both the G66U and G66C mutations cause the E22D mutation of the NS2A protein. Subsequently, the expression of NS1' protein, plaque size, replication ability, and virulence to mice of the three mutant strains were examined. The results showed that the three mutant viruses could not express NS1' protein, and their proliferation ability in nerve cells and virulence to mice were significantly reduced. In addition, the SA14(G66C) was less virulent than the other two mutated viruses. Our results indicate that only when G is the 66th nucleotide of NS2A, the JEV can produce NS1' protein, which affects the virulence.
Collapse
Affiliation(s)
- Ning Tan
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, China
| | - Chen Chen
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, China
| | - Yang Ren
- Department of Laboratory Medicine, Jintang First People's Hospital, West China Hospital Sichuan University JinTang Hospital, Chengdu, 610400, China
| | - Rong Huang
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, China
| | - Zhuang Zhu
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, China
| | - Kui Xu
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, China
| | - Xiaoyao Yang
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, China
| | - Jian Yang
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, China
| | - Lei Yuan
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, China.
| |
Collapse
|
3
|
Jablunovsky A, Narayanan A, Jose J. Identification of a critical role for ZIKV capsid α3 in virus assembly and its genetic interaction with M protein. PLoS Negl Trop Dis 2024; 18:e0011873. [PMID: 38166143 PMCID: PMC10786401 DOI: 10.1371/journal.pntd.0011873] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/12/2024] [Accepted: 12/19/2023] [Indexed: 01/04/2024] Open
Abstract
Flaviviruses such as Zika and dengue viruses are persistent health concerns in endemic regions worldwide. Efforts to combat the spread of flaviviruses have been challenging, as no antivirals or optimal vaccines are available. Prevention and treatment of flavivirus-induced diseases require a comprehensive understanding of their life cycle. However, several aspects of flavivirus biogenesis, including genome packaging and virion assembly, are not well characterized. In this study, we focused on flavivirus capsid protein (C) using Zika virus (ZIKV) as a model to investigate the role of the externally oriented α3 helix (C α3) without a known or predicted function. Alanine scanning mutagenesis of surface-exposed amino acids on C α3 revealed a critical CN67 residue essential for ZIKV virion production. The CN67A mutation did not affect dimerization or RNA binding of purified C protein in vitro. The virus assembly is severely affected in cells transfected with an infectious cDNA clone of ZIKV with CN67A mutation, resulting in a highly attenuated phenotype. We isolated a revertant virus with a partially restored phenotype by continuous passage of the CN67A mutant virus in Vero E6 cells. Sequence analysis of the revertant revealed a second site mutation in the viral membrane (M) protein MF37L, indicating a genetic interaction between the C and M proteins of ZIKV. Introducing the MF37L mutation on the mutant ZIKV CN67A generated a double-mutant virus phenotypically consistent with the isolated genetic revertant. Similar results were obtained with analogous mutations on C and M proteins of dengue virus, suggesting the critical nature of C α3 and possible C and M residues contributing to virus assembly in other Aedes-transmitted flaviviruses. This study provides the first experimental evidence of a genetic interaction between the C protein and the viral envelope protein M, providing a mechanistic understanding of the molecular interactions involved in the assembly and budding of Aedes-transmitted flaviviruses.
Collapse
Affiliation(s)
- Anastazia Jablunovsky
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Anoop Narayanan
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Joyce Jose
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
4
|
Gracias S, Chazal M, Decombe A, Unterfinger Y, Sogues A, Pruvost L, Robert V, Lacour SA, Lemasson M, Sourisseau M, Li Z, Richardson J, Pellegrini S, Decroly E, Caval V, Jouvenet N. Tick-borne flavivirus NS5 antagonizes interferon signaling by inhibiting the catalytic activity of TYK2. EMBO Rep 2023; 24:e57424. [PMID: 37860832 PMCID: PMC10702846 DOI: 10.15252/embr.202357424] [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: 05/02/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/21/2023] Open
Abstract
The mechanisms utilized by different flaviviruses to evade antiviral functions of interferons are varied and incompletely understood. Using virological approaches, biochemical assays, and mass spectrometry analyses, we report here that the NS5 protein of tick-borne encephalitis virus (TBEV) and Louping Ill virus (LIV), two related tick-borne flaviviruses, antagonize JAK-STAT signaling through interactions with the tyrosine kinase 2 (TYK2). Co-immunoprecipitation (co-IP) experiments, yeast gap-repair assays, computational protein-protein docking and functional studies identify a stretch of 10 residues of the RNA dependent RNA polymerase domain of tick-borne flavivirus NS5, but not mosquito-borne NS5, that is critical for interactions with the TYK2 kinase domain. Additional co-IP assays performed with several TYK2 orthologs reveal that the interaction is conserved across mammalian species. In vitro kinase assays show that TBEV and LIV NS5 reduce the catalytic activity of TYK2. Our results thus illustrate a novel mechanism by which viruses suppress the interferon response.
Collapse
Affiliation(s)
- Ségolène Gracias
- Virus Sensing and Signaling Unit, CNRS UMR3569, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Maxime Chazal
- Virus Sensing and Signaling Unit, CNRS UMR3569, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Alice Decombe
- AFMB UMR 7257, CNRS, Aix Marseille Université, Marseille, France
| | - Yves Unterfinger
- UMR1161 Virologie Laboratoire de Santé Animale, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Adrià Sogues
- Structural and Molecular Microbiology, VIB-VUB, Center for Structural Biology, Brussels, Belgium
| | - Lauryne Pruvost
- Virus Sensing and Signaling Unit, CNRS UMR3569, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Valentine Robert
- AFMB UMR 7257, CNRS, Aix Marseille Université, Marseille, France
| | - Sandrine A Lacour
- UMR1161 Virologie Laboratoire de Santé Animale, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Manon Lemasson
- UMR1161 Virologie Laboratoire de Santé Animale, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
- Phagos Pépinière Genopole Entreprise, Evry-Courcouronnes, France
| | - Marion Sourisseau
- UMR1161 Virologie Laboratoire de Santé Animale, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Zhi Li
- Unit of Cytokine Signaling, INSERM U122, Institut Pasteur, Paris, France
- Human Evolutionary Genetics Unit, CNRS UMR2000, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Jennifer Richardson
- UMR1161 Virologie Laboratoire de Santé Animale, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Sandra Pellegrini
- Unit of Cytokine Signaling, INSERM U122, Institut Pasteur, Paris, France
| | - Etienne Decroly
- AFMB UMR 7257, CNRS, Aix Marseille Université, Marseille, France
| | - Vincent Caval
- Virus Sensing and Signaling Unit, CNRS UMR3569, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Nolwenn Jouvenet
- Virus Sensing and Signaling Unit, CNRS UMR3569, Institut Pasteur, Université de Paris Cité, Paris, France
| |
Collapse
|
5
|
Virulence and Cross-Protection Conferred by an Attenuated Genotype I-Based Chimeric Japanese Encephalitis Virus Strain Harboring the E Protein of Genotype V in Mice. Microbiol Spectr 2022; 10:e0199022. [PMID: 36301111 PMCID: PMC9769820 DOI: 10.1128/spectrum.01990-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Japanese encephalitis virus (JEV) genotype V (GV) emerged in China in 2009, then South Korea, and has since spread to other regions in Asia and beyond, raising concern about its pathogenicity and the cross-protection offered by JEV vaccines against different genotypes. In this study, we replaced the structural proteins (C-prM-E) of an attenuated genotype I (GI) SD12-F120 strain with those of a virulent GV XZ0934 strain to construct a recombinant chimeric GI-GV JEV (JEV-GI/V) strain to determine the role of the structural proteins in virulence and cross-protection. The recombinant chimeric virus was highly neurovirulent and neuroinvasive in mice. This demonstrated the determinant role of the structural proteins in the virulence of the GV strain. Intracerebral or intraperitoneal inoculation of mice with JEV-GI/V-E5 harboring a combination of substitutions (N47K, L107F, E138K, H123R, and I176R) in E protein, but not mutants containing single substitution of these residues, resulted in decreased or disappeared mortality, suggesting that these residues synergistically, but not individually, played a role in determining the neurovirulence and neuroinvasiveness of the GV strain. Immunization of mice with attenuated strain JEV-GI/V-E5 provided complete protection and induced high neutralizing antibody titers against parental strain JEV-GI/V, but partial cross-protection and low cross-neutralizing antibodies titers against the heterologous GI and GIII strains in mice, suggesting the reduced cross-protection of JEV vaccines among different genotypes. Overall, these findings suggested the essential role of the structural proteins in determination of the virulence of GV strain, and highlighted the need for a novel vaccine against this newly emerged strain. IMPORTANCE The GV JEV showed an increase in epidemic areas, which exhibited higher pathogenicity in mice than the prevalent GI and GIII strains. We replaced a recombinant chimeric GI-GV JEV (JEV-GI/V) strain to determine the role of the structural proteins in virulence and cross-protection. It was found that the essential role of the structural proteins is to determinethe virulence of the GV strain. It is also suggested that there is reduced cross-protection of JEV vaccines among different genotypes, which provides basic data for subsequent JEV prevention, control, and new vaccine development.
Collapse
|
6
|
Isolation and Genetic Characterization of Japanese Encephalitis Virus Two Decades after Its Elimination in Singapore. Viruses 2022; 14:v14122662. [PMID: 36560666 PMCID: PMC9786948 DOI: 10.3390/v14122662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
Japanese encephalitis virus (JEV) is an important arbovirus in Asia that can cause serious neurological disease. JEV is transmitted by mosquitoes in an enzootic cycle involving porcine and avian reservoirs, in which humans are accidental, dead-end hosts. JEV is currently not endemic in Singapore, after pig farming was abolished in 1992; the last known human case was reported in 2005. However, due to its location along the East-Asian Australasian Flyway (EAAF), Singapore is vulnerable to JEV re-introduction from the endemic regions. Serological and genetic evidence in the last decade suggests JEV's presence in the local fauna. In the present study, we report the genetic characterization and the first isolation of JEV from 3214 mosquito pools consisting of 41,843 Culex mosquitoes, which were trapped from April 2014 to May 2021. The findings demonstrated the presence of genotype I of JEV (n = 10), in contrast to the previous reports of the presence of genotype II of JEV in Singapore. The genetic analyses also suggested that JEV has entered Singapore on several occasions and has potentially established an enzootic cycle in the local fauna. These observations have important implications in the risk assessment and the control of Japanese encephalitis in non-endemic countries, such as Singapore, that are at risk for JEV transmission.
Collapse
|
7
|
Davis EH, Wang B, White M, Huang YJS, Sarathy VV, Wang T, Bourne N, Higgs S, Barrett ADT. Impact of yellow fever virus envelope protein on wild-type and vaccine epitopes and tissue tropism. NPJ Vaccines 2022; 7:39. [PMID: 35322047 PMCID: PMC8942996 DOI: 10.1038/s41541-022-00460-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 02/16/2022] [Indexed: 12/04/2022] Open
Abstract
The envelope (E) protein of flaviviruses is functionally associated with viral tissue tropism and pathogenicity. For yellow fever virus (YFV), viscerotropic disease primarily involving the liver is pathognomonic for wild-type (WT) infection. In contrast, the live-attenuated vaccine (LAV) strain 17D does not cause viscerotropic disease and reversion to virulence is associated with neurotropic disease. The relationship between structure-function of the E protein for WT strain Asibi and its LAV derivative 17D strain is poorly understood; however, changes to WT and vaccine epitopes have been associated with changes in virulence. Here, a panel of Asibi and 17D infectious clone mutants were generated with single-site mutations at the one membrane residue and each of the eight E protein amino acid substitutions that distinguish the two strains. The mutants were characterized with respect to WT-specific and vaccine-specific monoclonal antibodies (mAbs) binding to virus plus binding of virus to brain, liver, and lung membrane receptor preparations (MRPs) generated from AG129 mice. This approach shows that amino acids in the YFV E protein domains (ED) I and II contain the WT E protein epitope, which overlap with those that mediate YFV binding to mouse liver. Furthermore, amino acids in EDIII associated with the vaccine epitope overlap with those that facilitate YFV binding mouse brain MRPs. Taken together, these data suggest that the YFV E protein is a key determinant in the phenotype of WT and 17D vaccine strains of YFV.
Collapse
Affiliation(s)
- Emily H Davis
- Department of Pathology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
- Sealy Institute for Vaccine Sciences, UTMB, Galveston, TX, USA
| | - Binbin Wang
- Department of Pathology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | | | - Yan-Jang S Huang
- Biosecurity Research Institute, Kansas State University, Manhattan, KS, USA
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
- Center on Emerging and Zoonotic Infectious Diseases, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Vanessa V Sarathy
- Department of Pathology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
- Sealy Institute for Vaccine Sciences, UTMB, Galveston, TX, USA
| | - Tian Wang
- Department of Pathology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
- Sealy Institute for Vaccine Sciences, UTMB, Galveston, TX, USA
| | - Nigel Bourne
- Sealy Institute for Vaccine Sciences, UTMB, Galveston, TX, USA
- Department of Pediatrics, UTMB, Galveston, TX, USA
| | - Stephen Higgs
- Biosecurity Research Institute, Kansas State University, Manhattan, KS, USA
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
- Center on Emerging and Zoonotic Infectious Diseases, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Alan D T Barrett
- Department of Pathology, University of Texas Medical Branch (UTMB), Galveston, TX, USA.
- Sealy Institute for Vaccine Sciences, UTMB, Galveston, TX, USA.
| |
Collapse
|
8
|
Khou C, Díaz-Salinas MA, da Costa A, Préhaud C, Jeannin P, Afonso PV, Vignuzzi M, Lafon M, Pardigon N. Comparative analysis of neuroinvasion by Japanese encephalitis virulent and vaccine viral strains in an in vitro model of human blood-brain barrier. PLoS One 2021; 16:e0252595. [PMID: 34086776 PMCID: PMC8177624 DOI: 10.1371/journal.pone.0252595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 03/16/2021] [Indexed: 11/18/2022] Open
Abstract
Japanese encephalitis virus (JEV) is the major cause of viral encephalitis in South East Asia. It has been suggested that, as a consequence of the inflammatory process during JEV infection, there is disruption of the blood-brain barrier (BBB) tight junctions that in turn allows the virus access to the central nervous system (CNS). However, what happens at early times of JEV contact with the BBB is poorly understood. In the present work, we evaluated the ability of both a virulent and a vaccine strain of JEV (JEV RP9 and SA14-14-2, respectively) to cross an in vitro human BBB model. Using this system, we demonstrated that both JEV RP9 and SA14-14-2 are able to cross the BBB without disrupting it at early times post viral addition. Furthermore, we find that almost 10 times more RP9 infectious particles than SA14-14 cross the model BBB, indicating this BBB model discriminates between the virulent RP9 and the vaccine SA14-14-2 strains of JEV. Beyond contributing to the understanding of early events in JEV neuroinvasion, we demonstrate this in vitro BBB model can be used as a system to study the viral determinants of JEV neuroinvasiveness and the molecular mechanisms by which this flavivirus crosses the BBB during early times of neuroinvasion.
Collapse
Affiliation(s)
- Cécile Khou
- Unité de Recherche et d’Expertise Environnement et Risques Infectieux, Groupe Arbovirus, Institut Pasteur, Paris, France
| | - Marco Aurelio Díaz-Salinas
- Unité de Recherche et d’Expertise Environnement et Risques Infectieux, Groupe Arbovirus, Institut Pasteur, Paris, France
| | - Anaelle da Costa
- Unité de Neuro-Immunologie Virale, Institut Pasteur, Paris, France
| | | | - Patricia Jeannin
- Unité d’Epidémiologie et Physiopathologie des Virus Oncogènes, Institut Pasteur, CNRS UMR 3569, Paris, France
| | - Philippe V. Afonso
- Unité d’Epidémiologie et Physiopathologie des Virus Oncogènes, Institut Pasteur, CNRS UMR 3569, Paris, France
| | - Marco Vignuzzi
- Unité des Populations Virales et Pathogenèse, Institut Pasteur, Paris, France
| | - Monique Lafon
- Unité de Neuro-Immunologie Virale, Institut Pasteur, Paris, France
| | - Nathalie Pardigon
- Unité de Recherche et d’Expertise Environnement et Risques Infectieux, Groupe Arbovirus, Institut Pasteur, Paris, France
- * E-mail:
| |
Collapse
|
9
|
Zika M Oligopeptide ZAMP Confers Cell Death-Promoting Capability to a Soluble Tumor-Associated Antigen through Caspase-3/7 Activation. Int J Mol Sci 2020; 21:ijms21249578. [PMID: 33339164 PMCID: PMC7765671 DOI: 10.3390/ijms21249578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 12/31/2022] Open
Abstract
Mosquito-borne Zika virus (ZIKV) is an emerging flavivirus of medical concern associated with neurological disorders. ZIKV utilizes apoptosis as a mechanism of cell killing. The structural M protein may play a role in flavivirus-induced apoptosis. The death-promoting capability of M has been restricted to an oligopeptide representing the residues M-32/40. Here, we evaluated the apoptosis inducing ability of the residues M-31/41 of ZIKV. The ZIKV M oligopeptide was associated to a soluble form of GFP (sGFP) and the resulting sGFP-M31/41 construct was assessed in Huh7 cells. Expression of sGFP-M31/41 can trigger apoptosis in Huh7 cells through caspase-3/7 activation. The translocation of sGFP-M31/41 in the endoplasmic reticulum was a prerequisite for apoptosis induction. The residues M-33/35/38 may play a critical role in the death-promoting activity of sGFP-M31/41. The effect of ZIKV M oligopeptide defined as ZAMP (for Zika Apoptosis M Peptide) on expression of a tumor-associated antigen was assayed on megakaryocyte-potentiating factor (MPF). Expression of MPF-ZAMP construct resulted in caspase-associated apoptosis activation in A549 and Huh7 cells. ZIKV has been proposed as an oncolytic virus for cancer therapy. The ability of the Zika M oligopeptide to confer death-promoting capability to MPF opens up attractive perspectives for ZAMP as an innovative anticancer agent.
Collapse
|
10
|
Adaptation of a live-attenuated genotype I Japanese encephalitis virus to vero cells is associated with mutations in structural protein genes. Virus Res 2020; 292:198256. [PMID: 33285172 DOI: 10.1016/j.virusres.2020.198256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/28/2020] [Accepted: 11/30/2020] [Indexed: 11/24/2022]
Abstract
The SD12-F120 is a live-attenuated genotype I strain of Japanese encephalitis virus (JEV) and was obtained by serial passage of wild-type strain SD12 on BHK-21 cells combined with multiple plaque purification and virulence selection in mice. The large scale production and vast clinical trials always demand ideal safety and efficacy profile of live-attenuated vaccines. In the present study, SD12-F120VC has undergone serial passaging of P1-P30 in WHO qualified Vero cells to assess the potential effect of adaptation to growth on Vero cells. The series of experiments showed that vaccine SD12-F120VC (Vero cell adapted) variants have consistently increased in peak virus titer compared to early passages and have good adaptation to growth in Vero cells. The animal experiments showed that Vero cell adapted SD12-F120VC variants have attenuation phenotype in suckling mice and the plaque morphology for all SD12-F120VC variants was small. Vaccination of mice with SD12-F120VC vaccine produced complete protection for homologous SD12 genotype I strain, but failed to give the complete protection of vaccinated mice against the challenge of heterologous N28 genotype III strain. In response to immunization of SD12-F120VC in mice, the neutralizing antibodies titer against homologous SD12-F120VC and SD12 (GI) was higher than heterologous N28 (GIII) strain. The prM protein has 6 amino acid substitutions, of which 5 amino acid changes were confined at the start of the pr domain in the ∼40 amino acids, and some mutations in the pr domain of prM might contribute to Vero cell adaptation. Our findings in this study are important for validation, evaluation and quality control study of live attenuated flaviviruses vaccines and show that Vero cells are a suitable substrate for the production of a safe and stable live-attenuated JEV vaccine.
Collapse
|
11
|
A Molecular Determinant of West Nile Virus Secretion and Morphology as a Target for Viral Attenuation. J Virol 2020; 94:JVI.00086-20. [PMID: 32269117 PMCID: PMC7307099 DOI: 10.1128/jvi.00086-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/29/2020] [Indexed: 12/22/2022] Open
Abstract
West Nile virus (WNV) is a worldwide (re)emerging mosquito-transmitted Flavivirus causing fatal neurological diseases in humans. However, no human vaccine has been yet approved. One of the most effective live-attenuated vaccines was empirically obtained by serial passaging of wild-type yellow fever Flavivirus. However, such an approach is not acceptable nowadays, and the development of a rationally designed vaccine is necessary. Generating molecular infectious clones and mutating specific residues known to be involved in Flavivirus virulence constitute a powerful tool to promote viral attenuation. WNV membrane glycoprotein is thought to carry such essential determinants. Here, we identified two residues of this protein whose substitutions are key to the full and stable attenuation of WNV in vivo, most likely through inhibition of secretion and possible alteration of morphology. Applied to other flaviviruses, this approach should help in designing new vaccines against these viruses, which are an increasing threat to global human health. West Nile virus (WNV), a member of the Flavivirus genus and currently one of the most common arboviruses worldwide, is associated with severe neurological disease in humans. Its high potential to reemerge and rapidly disseminate makes it a bona fide global public health problem. The surface membrane glycoprotein (M) has been associated with Flavivirus-induced pathogenesis. Here, we identified a key amino acid residue at position 36 of the M protein whose mutation impacts WNV secretion and promotes viral attenuation. We also identified a compensatory site at position M-43 whose mutation stabilizes M-36 substitution both in vitro and in vivo. Moreover, we found that introduction of the two mutations together confers a full attenuation phenotype and protection against wild-type WNV lethal challenge, eliciting potent neutralizing-antibody production in mice. Our study thus establishes the M protein as a new viral target for rational design of attenuated WNV strains. IMPORTANCE West Nile virus (WNV) is a worldwide (re)emerging mosquito-transmitted Flavivirus causing fatal neurological diseases in humans. However, no human vaccine has been yet approved. One of the most effective live-attenuated vaccines was empirically obtained by serial passaging of wild-type yellow fever Flavivirus. However, such an approach is not acceptable nowadays, and the development of a rationally designed vaccine is necessary. Generating molecular infectious clones and mutating specific residues known to be involved in Flavivirus virulence constitute a powerful tool to promote viral attenuation. WNV membrane glycoprotein is thought to carry such essential determinants. Here, we identified two residues of this protein whose substitutions are key to the full and stable attenuation of WNV in vivo, most likely through inhibition of secretion and possible alteration of morphology. Applied to other flaviviruses, this approach should help in designing new vaccines against these viruses, which are an increasing threat to global human health.
Collapse
|
12
|
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
|
13
|
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
|
14
|
Li L, Collins ND, Widen SG, Davis EH, Kaiser JA, White MM, Greenberg MB, Barrett ADT, Bourne N, Sarathy VV. Attenuation of Zika Virus by Passage in Human HeLa Cells. Vaccines (Basel) 2019; 7:vaccines7030093. [PMID: 31434319 PMCID: PMC6789458 DOI: 10.3390/vaccines7030093] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/14/2019] [Accepted: 08/16/2019] [Indexed: 12/14/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne Flavivirus. Previous studies have shown that mosquito-transmitted flaviviruses, including yellow fever, Japanese encephalitis, and West Nile viruses, could be attenuated by serial passaging in human HeLa cells. Therefore, it was hypothesized that wild-type ZIKV would also be attenuated after HeLa cell passaging. A human isolate from the recent ZIKV epidemic was subjected to serial HeLa cell passaging, resulting in attenuated in vitro replication in both Vero and A549 cells. Additionally, infection of AG129 mice with 10 plaque forming units (pfu) of wild-type ZIKV led to viremia and mortality at 12 days, whereas infection with 103 pfu of HeLa-passage 6 (P6) ZIKV led to lower viremia, significant delay in mortality (median survival: 23 days), and increased cytokine and chemokine responses. Genomic sequencing of HeLa-passaged virus identified two amino acid substitutions as early as HeLa-P3: pre-membrane E87K and nonstructural protein 1 R103K. Furthermore, both substitutions were present in virus harvested from HeLa-P6-infected animal tissue. Together, these data show that, similarly to other mosquito-borne flaviviruses, ZIKV is attenuated following passaging in HeLa cells. This strategy can be used to improve understanding of substitutions that contribute to attenuation of ZIKV and be applied to vaccine development across multiple platforms.
Collapse
Affiliation(s)
- Li Li
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Natalie D Collins
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Viral Disease Branch, Walter Reed Army Institute for Research, Silver Spring, MD 20910, USA
| | - Steven G Widen
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Emily H Davis
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jaclyn A Kaiser
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mellodee M White
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - M Banks Greenberg
- Department of Pediatrics, 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
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Nigel Bourne
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Vanessa V Sarathy
- 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.
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
15
|
Review of Emerging Japanese Encephalitis Virus: New Aspects and Concepts about Entry into the Brain and Inter-Cellular Spreading. Pathogens 2019; 8:pathogens8030111. [PMID: 31357540 PMCID: PMC6789543 DOI: 10.3390/pathogens8030111] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
Japanese encephalitis virus (JEV) is an emerging flavivirus of the Asia-Pacific region. More than two billion people live in endemic or epidemic areas and are at risk of infection. Recently, the first autochthonous human case was recorded in Africa, and infected birds have been found in Europe. JEV may spread even further to other continents. The first section of this review covers established and new information about the epidemiology of JEV. The subsequent sections focus on the impact of JEV on humans, including the natural course and immunity. Furthermore, new concepts are discussed about JEV’s entry into the brain. Finally, interactions of JEV and host cells are covered, as well as how JEV may spread in the body through latently infected immune cells and cell-to-cell transmission of virions or via other infectious material, including JEV genomic RNA.
Collapse
|
16
|
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
|
17
|
Gamma-interferon exerts a critical early restriction on replication and dissemination of yellow fever virus vaccine strain 17D-204. NPJ Vaccines 2018; 3:5. [PMID: 29387474 PMCID: PMC5780476 DOI: 10.1038/s41541-017-0039-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/27/2017] [Accepted: 11/30/2017] [Indexed: 11/11/2022] Open
Abstract
Live attenuated viruses are historically among the most effective viral vaccines. Development of a safe vaccine requires the virus to be less virulent, a phenotype that is historically arrived by empirical evaluation often leaving the mechanisms of attenuation unknown. The yellow fever virus 17D live attenuated vaccine strain has been developed as a delivery vector for heterologous antigens; however, the mechanisms of attenuation remain elusive. The successful and safe progress of 17D as a vaccine vector and the development of live attenuated vaccines (LAVs) to related flaviviruses requires an understanding of the molecular mechanisms leading to attenuation. Using subcutaneous infection of interferon-deficient mouse models of wild type yellow fever virus (WT YFV) pathogenesis and 17D-mediated immunity, we found that, in the absence of type I IFN (IFN-α/β), type II interferon (IFN-γ) restricted 17D replication, but not that of WT YFV, by 1–2 days post-infection. In this context, IFN-γ responses protected 17D-infected animals from mortality, largely restricted the virus to lymphoid organs, and eliminated viscerotropic disease signs such as steatosis in the liver and inflammatory cell infiltration into the spleen. However, WT YFV caused a disseminated infection, gross liver pathology, and rapid death of the animals. In vitro, IFN-γ treatment of myeloid cells suppressed the replication of 17D significantly more than that of WT YFV, suggesting a direct differential effect on 17D virus replication. Together these data indicate that an important mechanism of 17D attenuation in vivo is increased sensitivity to IFN-γ stimulated responses elicited early after infection. The interferon gamma protein may play a key role in preventing yellow fever vaccine 17D from causing virus-like disease in recipients. The highly effective 17D vaccine is a less virulent form of the virus, but can induce severe disease in rare cases. A research group from the University of Pittsburgh, led by William Klimstra, investigated the impact of the vaccine on mice, as the mechanism by which hosts defend against its disease-causing potential is not fully understood. They found that interferon gamma restricted the replication and spread of the attenuated virus (the vaccine) but not its natural form. This study helps to inform efforts to improve the safety of the 17D vaccine, as well as the other vaccines that use it as a template for prophylaxis against other pathogens.
Collapse
|
18
|
Kaiser JA, Wang T, Barrett AD. Virulence determinants of West Nile virus: how can these be used for vaccine design? Future Virol 2017; 12:283-295. [PMID: 28919920 DOI: 10.2217/fvl-2016-0141] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/14/2017] [Indexed: 12/12/2022]
Abstract
West Nile virus (WNV), a neurotropic mosquito-borne flavivirus, has become endemic in the USA and parts of Europe since 1999. There is no licensed WNV vaccine for humans. Considering the robust immunity from immunization with live, attenuated vaccines, a live WNV vaccine is an ideal platform for disease control. Animal and mosquito studies have identified a number of candidate attenuating mutations, including the structural proteins premembrane/membrane and envelope, and the nonstructural proteins NS1, NS2A, NS3, NS4A, NS4B and NS5, and the 3' UTR. Many of the mutations that have been examined attenuate WNV using different mechanisms, thus providing a greater understanding of WNV virulence while also identifying specific mutations as candidates to include in a WNV live vaccine.
Collapse
Affiliation(s)
- Jaclyn A Kaiser
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Alan Dt Barrett
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
19
|
Lu CY, Hour MJ, Wang CY, Huang SH, Mu WX, Chang YC, Lin CW. Single-Round Infectious Particle Antiviral Screening Assays for the Japanese Encephalitis Virus. Viruses 2017; 9:v9040076. [PMID: 28394283 PMCID: PMC5408682 DOI: 10.3390/v9040076] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 12/13/2022] Open
Abstract
Japanese Encephalitis virus (JEV) is a mosquito-borne flavivirus with a positive-sense single-stranded RNA genome that contains a big open reading frame (ORF) flanked by 5′- and 3′- untranslated regions (UTRs). Nearly 30,000 JE cases with 10,000 deaths are still annually reported in East Asia. Although the JEV genotype III vaccine has been licensed, it elicits a lower protection against other genotypes. Moreover, no effective treatment for a JE case is developed. This study constructed a pBR322-based and cytomegaloviruses (CMV) promoter-driven JEV replicon for the production of JEV single-round infectious particles (SRIPs) in a packaging cell line expressing viral structural proteins. Genetic instability of JEV genome cDNA in the pBR322 plasmid was associated with the prokaryotic promoter at 5′ end of the JEV genome that triggers the expression of the structural proteins in E. coli. JEV structural proteins were toxic E. coli, thus the encoding region for structural proteins was replaced by a reporter gene (enhanced green fluorescent protein, EGFP) that was in-frame fused with the first eight amino acids of the C protein at N-terminus and the foot-and-mouth disease virus (FMDV) 2A peptide at C-terminus in a pBR322-based JEV-EGFP replicon. JEV-EGFP SRIPs generated from JEV-EGFP replicon-transfected packaging cells displayed the infectivity with cytopathic effect induction, self-replication of viral genomes, and the expression of EGFP and viral proteins. Moreover, the combination of JEV-EGFP SRIP plus flow cytometry was used to determine the half maximal inhibitory concentration (IC50) values of antiviral agents according to fluorescent intensity and positivity of SRIP-infected packaging cells post treatment. MJ-47, a quinazolinone derivative, significantly inhibited JEV-induced cytopathic effect, reducing the replication and expression of JEV-EGFP replicon in vitro. The IC50 value of 6.28 µM for MJ-47 against JEV was determined by the assay of JEV-EGFP SRIP infection in packaging cells plus flow cytometry that was more sensitive, effective, and efficient compared to the traditional plaque assay. Therefore, the system of JEV-EGFP SRIPs plus flow cytometry was a rapid and reliable platform for screening antiviral agents and evaluating antiviral potency.
Collapse
Affiliation(s)
- Chien-Yi Lu
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan.
| | - Mann-Jen Hour
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan.
| | - Ching-Ying Wang
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan.
| | - Su-Hua Huang
- Department of Biotechnology, Asia University, Taichung 41354, Taiwan.
| | - Wen-Xiang Mu
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan.
| | - Yu-Chun Chang
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan.
| | - Cheng-Wen Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan.
- Department of Biotechnology, Asia University, Taichung 41354, Taiwan.
| |
Collapse
|