1
|
Zhang T, Chen Z, Xie L, Xu R, Chen L, Jia T, Shi W, Wang Y, Song Y, Han Q, Xia X, Yuan T, Zhang J. A fusion protein of vimentin with Fc fragment inhibits Japanese encephalitis virus replication. Front Vet Sci 2024; 11:1368725. [PMID: 38500602 PMCID: PMC10944967 DOI: 10.3389/fvets.2024.1368725] [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: 01/11/2024] [Accepted: 02/21/2024] [Indexed: 03/20/2024] Open
Abstract
Japanese encephalitis virus (JEV), a member of the Flaviviridae family and a flavivirus, is known to induce acute encephalitis. Vimentin protein has been identified as a potential receptor for JEV, engaging in interactions with the viral membrane protein. The Fc fragment, an integral constituent of immunoglobulins, plays a crucial role in antigen recognition by dendritic cells (DCs) or phagocytes, leading to subsequent antigen presentation, cytotoxicity, or phagocytosis. In this study, we fused the receptor of JEV vimentin with the Fc fragment of IgG and expressed the resulting vimentin-Fc fusion protein in Escherichia coli. Pull-down experiments demonstrated the binding ability of the vimentin-Fc fusion protein to JEV virion in vitro. Additionally, we conducted inhibition assays at the cellular level, revealing the ability of vimentin-Fc protein suppressing JEV replication, it may be a promising passive immunotherapy agent for JEV. These findings pave the way for potential therapeutic strategies against JEV.
Collapse
Affiliation(s)
- Taoping Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
- Yunnan Provincial Key Laboratory of Clinical Virology, The First People's Hospital of Yunnan Province, Kunming, China
- Yunnan Province Clinical Research Center for Gynecological and Obstetric Disease, Kunming, China
| | - Zhixin Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Lyu Xie
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ruixian Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Lu Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Ting Jia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Wengang Shi
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Yongbo Wang
- Department of Clinical Laboratory, The First People's Hospital of Yunnan Province, Kunming, China
| | - Yuzhu Song
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Qinqin Han
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Xueshan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Tao Yuan
- Yunnan Provincial Key Laboratory of Clinical Virology, The First People's Hospital of Yunnan Province, Kunming, China
- Department of Gynecology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Jinyang Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
- Yunnan Province Clinical Research Center for Gynecological and Obstetric Disease, Kunming, China
| |
Collapse
|
2
|
Lee E, Kim M, Kim YB. Attenuated Chimeric GI/GIII Vaccine Candidate against Japanese Encephalitis Virus. Vaccines (Basel) 2023; 11:1827. [PMID: 38140231 PMCID: PMC10747704 DOI: 10.3390/vaccines11121827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Japanese encephalitis (JE) is a very severe disease characterized by high fatality rates and the development of permanent behavioral, psychiatric, and neurological sequelae among survivors. Japanese encephalitis virus (JEV), a flavivirus, is responsible for JE. In Asia, Genotype I (GI) has emerged as the dominant strain, replacing Genotype III (GIII). However, no clinically approved drug is available to treat JEV infection, and currently available commercial vaccines derived from JEV GIII strains provide only partial protection against GI. Utilizing a reverse genetics system, this study attempted to produce a novel chimeric JEV strain with high efficacy against JEV GI. Accordingly, a GI/GIII intertypic recombinant strain, namely SA14-GI env, was generated by substituting the E region of the GIII SA14-14-2 strain with that of the GI strain, K05GS. The neurovirulence of the mutant virus was significantly reduced in mice. Analysis of the immunogenicity of the chimeric virus revealed that it induced neutralizing antibodies against JEV GI in mice, and the protective efficacy of SA14-GI env was higher than that of SA14-14-2. These findings suggest that SA14-GI env may be a safe and effective live-attenuated vaccine candidate against JEV GI.
Collapse
Affiliation(s)
| | | | - Young Bong Kim
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Republic of Korea; (E.L.); (M.K.)
| |
Collapse
|
3
|
Xia Q, Yang Y, Zhang Y, Zhou L, Ma X, Xiao C, Zhang J, Li Z, Liu K, Li B, Shao D, Qiu Y, Wei J, Ma Z. Shift in dominant genotypes of Japanese encephalitis virus and its impact on current vaccination strategies. Front Microbiol 2023; 14:1302101. [PMID: 38045034 PMCID: PMC10690641 DOI: 10.3389/fmicb.2023.1302101] [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: 09/26/2023] [Accepted: 11/01/2023] [Indexed: 12/05/2023] Open
Abstract
Japanese encephalitis (JE) is a zoonotic ailment from the Japanese encephalitis virus (JEV). JEV belongs to the flavivirus genus and is categorized into a solitary serotype consisting of five genetically diverse genotypes (I, II, III, IV, and V). The JEV genotype III (GIII) was the prevailing strain responsible for multiple outbreaks in countries endemic to JEV until 1990. In recent years, significant improvements have occurred in the epidemiology of JE, encompassing the geographical expansion of the epidemic zone and the displacement of prevailing genotypes. The dominant genotype of the JEV has undergone a progressive shift from GIII to GI due to variations in its adaptability within avian populations. From 2021 to 2022, Australia encountered an epidemic of viral encephalitis resulting from infection with the GIV JEV pathogen. The current human viral encephalitis caused by GIV JEV is the initial outbreak since its initial discovery in Indonesia during the late 1970s. Furthermore, following a time frame of 50 years, the detection and isolation of GV JEV have been reported in Culex mosquitoes across China and South Korea. Evidence suggests that the prevalence of GIV and GV JEV epidemic regions may be on the rise, posing a significant threat to public safety and the sustainable growth of animal husbandry. The global approach to preventing and managing JE predominantly revolves around utilizing the GIII strain vaccine for vaccination purposes. Nevertheless, research has demonstrated that the antibodies generated by the GIII strain vaccine exhibit limited capacity to neutralize the GI and GV strains. Consequently, these antibodies cannot protect against JEV challenge caused by animal GI and GV strains. The limited cross-protective and neutralizing effects observed between various genotypes may be attributed to the low homology of the E protein with other genotypes. In addition, due to the GIV JEV outbreak in Australia, further experiments are needed to evaluate the protective efficiency of the current GIII based JE vaccine against GIV JEV. The alteration of the prevailing genotype of JEV and the subsequent enlargement of the geographical extent of the epidemic have presented novel obstacles in JE prevention and control. This paper examines the emerging features of the JE epidemic in recent years and the associated problems concerning prevention and control.
Collapse
Affiliation(s)
- Qiqi Xia
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yang Yang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yan Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Lujia Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Xiaochun Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Changguang Xiao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Junjie Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Zongjie Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Ke Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Beibei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Donghua Shao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yafeng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Zhiyong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
4
|
Dong N, Zhang X, Zhang H, Zheng J, Qiu Y, Li Z, Li B, Liu K, Shao D, Ma Z, Wei J. Genotype Change in Circulating JEV Strains in Fujian Province, China. Viruses 2023; 15:1822. [PMID: 37766229 PMCID: PMC10536422 DOI: 10.3390/v15091822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Japanese encephalitis (JE), found in pigs, is a serious mosquito-borne zoonotic infectious disease caused by the Japanese encephalitis virus (JEV). JEV is maintained in an enzootic cycle between mosquitoes and amplifying vertebrate hosts, mainly pigs and wading birds. It is transmitted to humans through the bite of an infected mosquito, allowing the pathogen to spread and cause disease epidemics. However, there is little research on JEV genotype variation in mosquitoes and pigs in Fujian province. Previous studies have shown that the main epidemic strain of JEV in Fujian Province is genotype III. In this study, a survey of mosquito species diversity in pig farms and molecular evolutionary analyses of JEV were conducted in Fujian, China, in the summer of 2019. A total of 19,177 mosquitoes were collected at four sites by UV trap. Four genera were identified, of which the Culex tritaeniorhynchus was the most common mosquito species, accounting for 76.4% of the total (14,651/19,177). Anopheles sinensi (19.25%, 3691/19,177) was the second largest species. High mosquito infection rateswere an important factor in the outbreak. The captured mosquito samples were milled and screened with JEV-specific primers. Five viruses were isolated, FJ1901, FJ1902, FJ1903, FJ1904, and FJ1905. Genetic affinity was determined by analyzing the envelope (E) gene variants. The results showed that they are JEV gene type I and most closely related to the strains SH-53 and SD0810. In this study, it was found through genetic evolution analysis that the main epidemic strain of JE in pig farms changed from gene type III to gene type I. Compared with the SH-53 and SD0810 strains, we found no change in key sites related to antigenic activity and neurovirulence of JEV in Fujian JEV and pig mosquito strains, respectively. The results of the study provide basic data for analyzing the genotypic shift of JEV in Fujian Province and support the prevention and control of JEV.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Zhiyong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (X.Z.); (H.Z.); (J.Z.); (Y.Q.); (Z.L.); (B.L.); (K.L.); (D.S.)
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (X.Z.); (H.Z.); (J.Z.); (Y.Q.); (Z.L.); (B.L.); (K.L.); (D.S.)
| |
Collapse
|
5
|
Wu X, Huang S, Wang M, Chen S, Liu M, Zhu D, Zhao X, Wu Y, Yang Q, Zhang S, Huang J, Ou X, Zhang L, Liu Y, Yu Y, Gao Q, Mao S, Sun D, Tian B, Yin Z, Jing B, Cheng A, Jia R. A novel live attenuated duck Tembusu virus vaccine targeting N7 methyltransferase protects ducklings against pathogenic strains. Vet Res 2023; 54:47. [PMID: 37308988 DOI: 10.1186/s13567-023-01170-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 03/28/2023] [Indexed: 06/14/2023] Open
Abstract
Duck Tembusu virus (DTMUV), an emerging pathogenic flavivirus, causes markedly decreased egg production in laying duck and neurological dysfunction and death in ducklings. Vaccination is currently the most effective means for prevention and control of DTMUV. In previous study, we have found that methyltransferase (MTase) defective DTMUV is attenuated and induces a higher innate immunity. However, it is not clear whether MTase-deficient DTMUV can be used as a live attenuated vaccine (LAV). In this study, we investigated the immunogenicity and immunoprotection of N7-MTase defective recombinant DTMUV K61A, K182A and E218A in ducklings. These three mutants were highly attenuated in both virulence and proliferation in ducklings but still immunogenic. Furthermore, a single-dose immunization with K61A, K182A or E218A could induce robust T cell responses and humoral immune responses, which could protect ducks from the challenge of a lethal-dose of DTMUV-CQW1. Together, this study provides an ideal strategy to design LAVs for DTMUV by targeting N7-MTase without changing the antigen composition. This attenuated strategy targeting N7-MTase may apply to other flaviviruses.
Collapse
Affiliation(s)
- Xuedong Wu
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shanzhi Huang
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Mingshu Wang
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China
| | - Shun Chen
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China
| | - Mafeng Liu
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China
| | - Dekang Zhu
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China
| | - Xinxin Zhao
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China
| | - Ying Wu
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China
| | - Qiao Yang
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China
| | - Shaqiu Zhang
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China
| | - Juan Huang
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China
| | - Xumin Ou
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China
| | - Ling Zhang
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yunya Liu
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yanling Yu
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Qun Gao
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Sai Mao
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Di Sun
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Bin Tian
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhongqiong Yin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China
| | - Bo Jing
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China
| | - Anchun Cheng
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China.
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China.
| | - Renyong Jia
- Research Centre of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Chengdu, 611130, China.
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, 611130, China.
| |
Collapse
|
6
|
Abstract
RNA viruses are not only reported for viral pandemics but also as important agents for emerging/re-emerging diseases. Japanese encephalitis virus (JEV) is reported to cause epidemics of encephalitis in Southeast Asia, India, Korea, China, and Indonesia. In addition, several reports show that JEV has spread to new populations beyond these geographical regions. The disease mostly affects children with a mortality rate up to 30%. In peridomestic settings, pigs are reported as amplifiers of JEV transmission and aquatic birds as maintenance hosts of the virus. The Culex mosquito is the vector for transmission of JEV. This virus is a member of the family Flaviviridae and has a single-stranded positive-sense RNA virus. Five different genotypes (G-I to G-V) of JEV have been reported. Four different kinds of vaccines have been produced to prevent JEV infection. However, there is no FDA-approved antiviral drug available for JEV. How to cite this article: Mehta A, Singh R, Mani VE, Poddar B. Japanese B Encephalitis. Indian J Crit Care Med 2021;25(Suppl 2):S171-S174.
Collapse
Affiliation(s)
- Anita Mehta
- Department of Pediatric Baba Raghav Das Medical College, Gorakhpur, Uttar Pradesh, India
| | | | - Vinita E Mani
- Department of Neurology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Banani Poddar
- Department of Critical Care Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| |
Collapse
|
7
|
Zhou D, Pei C, Liu Z, Yang K, Li Q, Chen H, Cao S, Song Y. Identification of a protective epitope in Japanese encephalitis virus NS1 protein. Antiviral Res 2020; 182:104930. [PMID: 32898585 DOI: 10.1016/j.antiviral.2020.104930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 08/23/2020] [Accepted: 09/02/2020] [Indexed: 12/16/2022]
Abstract
Japanese encephalitis virus (JEV) is one of the most important culex transmitted-flaviviruses, which can cause encephalitis in humans. Although non-structural protein 1 (NS1) of JEV does not stimulate neutralizing antibodies, this protein can provide high immunoprotection in vivo. The protective epitopes and the protective mechanism of NS1 still remain unclear. In this study, we generated five different monoclonal antibodies (mAbs) targeting the NS1 protein of JEV. In vitro experiments revealed that none of these five antibodies neutralized the JEV infection. In mouse protection studies, one of these mAbs, designated 2B8, provided a therapeutic effect against JEV lethal challenge (70% survival rate). Using peptide mapping analysis, we found that mAb 2B8 reacted with the epitope 225PETHTLWGD233 in the NS1 protein, in which any mutations among amino acid residues T228, H229, L231 or W232 could cause binding failure of 2B8 to the NS1 protein. Furthermore, mice immunized with KLH-polypeptide (225PETHTLWGD233) showed reduced mortality following JEV challenge. Collectively, we found a new protective epitope in the JEV NS1 protein. These results may facilitate the development of therapeutic agent and subunit-based vaccines based on the NS1 protein.
Collapse
Affiliation(s)
- Dengyuan Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chao Pei
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhaoxia Liu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Kelu Yang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Qiuyan Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shengbo Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yunfeng Song
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
8
|
Steffen T, Hassert M, Hoft SG, Stone ET, Zhang J, Geerling E, Grimberg BT, Roberts MS, Pinto AK, Brien JD. Immunogenicity and Efficacy of a Recombinant Human Adenovirus Type 5 Vaccine against Zika Virus. Vaccines (Basel) 2020; 8:vaccines8020170. [PMID: 32272595 PMCID: PMC7349816 DOI: 10.3390/vaccines8020170] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/26/2020] [Accepted: 03/29/2020] [Indexed: 12/20/2022] Open
Abstract
Zika virus (ZIKV) is a significant public health concern due to the pathogen's ability to be transmitted by either mosquito bite or sexual transmission, allowing spread to occur throughout the world. The potential consequences of ZIKV infection to human health, specifically neonates, necessitates the development of a safe and effective Zika virus vaccine. Here, we developed an intranasal Zika vaccine based upon the replication-deficient human adenovirus serotype 5 (hAd5) expressing ZIKV pre-membrane and envelope protein (hAd5-ZKV). The hAd5-ZKV vaccine is able to induce both cell-mediated and humoral immune responses to ZIKV epitopes. Importantly, this vaccine generated CD8+ T cells specific for a dominant ZIKV T cell epitope and is shown to be protective against a ZIKV challenge by using a pre-clinical model of ZIKV disease. We also demonstrate that the vaccine expresses pre-membrane and envelope protein in a confirmation recognized by ZIKV experienced individuals. Our studies demonstrate that this adenovirus-based vaccine expressing ZIKV proteins is immunogenic and protective in mice, and it encodes ZIKV proteins in a conformation recognized by the human antibody repertoire.
Collapse
Affiliation(s)
- Tara Steffen
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 63103, USA; (T.S.); (M.H.); (S.G.H.); (E.T.S.); (E.G.)
| | - Mariah Hassert
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 63103, USA; (T.S.); (M.H.); (S.G.H.); (E.T.S.); (E.G.)
| | - Stella G. Hoft
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 63103, USA; (T.S.); (M.H.); (S.G.H.); (E.T.S.); (E.G.)
| | - E. Taylor Stone
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 63103, USA; (T.S.); (M.H.); (S.G.H.); (E.T.S.); (E.G.)
| | - Jianfeng Zhang
- Altimmune, Inc., Gaithersburg, MD 20878, USA; (J.Z.); (M.S.R.)
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 63103, USA; (T.S.); (M.H.); (S.G.H.); (E.T.S.); (E.G.)
| | - Brian T. Grimberg
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA;
| | - M. Scot Roberts
- Altimmune, Inc., Gaithersburg, MD 20878, USA; (J.Z.); (M.S.R.)
| | - Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 63103, USA; (T.S.); (M.H.); (S.G.H.); (E.T.S.); (E.G.)
- Correspondence: (A.K.P.); (J.D.B.)
| | - James D. Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 63103, USA; (T.S.); (M.H.); (S.G.H.); (E.T.S.); (E.G.)
- Correspondence: (A.K.P.); (J.D.B.)
| |
Collapse
|
9
|
Nath B, Vandna, Saini HM, Prasad M, Kumar S. Evaluation of Japanese encephalitis virus E and NS1 proteins immunogenicity using a recombinant Newcastle disease virus in mice. Vaccine 2020; 38:1860-1868. [PMID: 31955960 DOI: 10.1016/j.vaccine.2019.11.088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/19/2019] [Accepted: 11/25/2019] [Indexed: 01/04/2023]
Abstract
Japanese encephalitis (JE) is the most important cause of acute encephalitis syndrome (AES). Japanese encephalitis virus (JEV), the prototype member of the JE serocomplex, belongs to the genus Flavivirus. The immunogenic proteins envelope (E) and non-structural protein 1 (NS1) of JEV are widely explored for the development of vaccines and diagnostics against JEV. However, there are underlying concerns such as the risk of reversion of live-attenuated vaccines to high virulence, the incomplete inactivation of pathogens in inactivated vaccines and partial vaccine coverage. Newcastle disease virus (NDV) is an efficient viral vaccine vector to express several human and animal immunogenic proteins. In the present study, we have developed a recombinant NDV (rNDV), individually expressing the E and NS1 proteins of JEV (rNDV-Ejev and rNDV-NS1jev). The recovered rNDV-Ejev and rNDV-NS1jev were characterized in 9-day-old SPF embryonated chicken eggs and in cell culture. The vaccination of rNDV-Ejev and rNDV-NS1jev showed effective immunity against JEV upon intranasal immunization in BALB/c mice. The rNDVs vaccination produced effective neutralization antibody titers against both NDV and JEV. The cytokine profiling of the vaccinated mice showed an effective Th1 and Th2 mediated immune response. The study also provided an insight that E, when used in combination with NS1 could reduce the efficacy of only E based immunization in mice. Our results suggested rNDV-Ejev to be a promising live viral vectored vaccine against JEV. This study implies an alternative and economical strategy for the development of a recombinant vaccine against JEV.
Collapse
Affiliation(s)
- Barnali Nath
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Vandna
- Center for Medical Biotechnology, M.D. University, Rohtak 124001, Haryana, India
| | - Hari Mohan Saini
- Center for Medical Biotechnology, M.D. University, Rohtak 124001, Haryana, India
| | - Minakshi Prasad
- College of Veterinary Sciences, Lala Lajpat Rai University of Veterinary & Animal Sciences, Hisar 125004, Haryana, India
| | - Sachin Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India.
| |
Collapse
|
10
|
Turtle L, Brindle HE, Schluter WW, Faragher B, Rayamajhi A, Bohara R, Gurung S, Shakya G, Yoksan S, Dixit S, Rajbhandari R, Paudel B, Adhikari S, Solomon T, Griffiths MJ. Low population Japanese encephalitis virus (JEV) seroprevalence in Udayapur district, Nepal, three years after a JE vaccination programme: A case for further catch up campaigns? PLoS Negl Trop Dis 2019; 13:e0007269. [PMID: 30986252 PMCID: PMC6483279 DOI: 10.1371/journal.pntd.0007269] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 04/25/2019] [Accepted: 02/27/2019] [Indexed: 02/07/2023] Open
Abstract
The live attenuated Japanese encephalitis (JE) vaccine SA14-14-2 has been used in Nepal for catch-up campaigns and is now included in the routine immunisation schedule. Previous studies have shown good vaccine efficacy after one dose in districts with a high incidence of JE. The first well-documented dengue outbreak occurred in Nepal in 2006 with ongoing cases now thought to be secondary to migration from India. Previous infection with dengue virus (DENV) partially protects against JE and might also influence serum neutralising antibody titres against JEV. This study aimed to determine whether serum anti-JEV neutralisation titres are: 1. maintained over time since vaccination, 2. vary with historic local JE incidence, and 3. are associated with DENV neutralising antibody levels. We conducted a cross-sectional study in three districts of Nepal: Banke, Rupandehi and Udayapur. Udayapur district had been vaccinated against JE most recently (2009), but had been the focus of only one campaign, compared with two in Banke and three in Rupandehi. Participants answered a short questionnaire and serum was assayed for anti-JEV and anti-DENV IgM and IgG (by ELISA) and 50% plaque reduction neutralisation titres (PRNT50) against JEV and DENV serotypes 1–4. A titre of ≥1:10 was considered seropositive to the respective virus. JEV neutralising antibody seroprevalence (PRNT50 ≥ 1:10) was 81% in Banke and Rupandehi, but only 41% in Udayapur, despite this district being vaccinated more recently. Sensitivity of ELISA for both anti-JEV and anti-DENV antibodies was low compared with PRNT50. DENV neutralising antibody correlated with the JEV PRNT50 ≥1:10, though the effect was modest. IgM (indicating recent infection) against both viruses was detected in a small number of participants. We also show that DENV IgM is present in Nepali subjects who have not travelled to India, suggesting that DENV may have become established in Nepal. We therefore propose that further JE vaccine campaigns should be considered in Udayapur district, and similar areas that have had fewer vaccination campaigns. In Nepal, immunisation using a live attenuated vaccine is given against Japanese encephalitis (JE), caused by the mosquito-transmitted JE virus (JEV). JE immunisation has taken place via catch-up campaigns and is now part of the routine immunisation programme. Although previous studies have shown good vaccine efficacy in areas where there is a lot of natural exposure to the virus (high endemicity), it is suggested that the efficacy may wane in areas where transmission is lower. Dengue virus (DENV) belongs to the same family and genus as JEV. Previous infection with DENV may also influence the immune response to JEV. Therefore, we conducted a cross-sectional study in Nepal to measure immunity to JE, in districts of differing historic JE incidence, and time from JE vaccination. This showed that neutralising antibody to JEV was found more frequently in districts which had been the subject of more vaccination campaigns, rather than in the most recently vaccinated district. In addition, we cannot rule out a role for natural exposure to JEV in maintaining higher antibody levels. Additionally, the study showed that previous exposure to DENV was positively associated with an immune response to JEV, though this effect was modest. We conclude that there is a need to consider further JE vaccine catch up campaigns in some areas especially given that we could detect JEV IgM, indicating ongoing transmission. We show that ELISA yielded many false negative results for exposure to JEV or vaccination, when compared with neutralising antibody. We also identified some individuals during the course of the study with DENV IgM in their blood, but with no history of travel to India. This suggests that DENV may have become established in some areas of Nepal.
Collapse
Affiliation(s)
- Lance Turtle
- Institute of Infection and Global Health and National Institute for Health Research Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, United Kingdom.,Royal Liverpool and Broadgreen University Hospitals, members of Liverpool Health Partners, Liverpool, United Kingdom
| | - Hannah E Brindle
- Institute of Infection and Global Health and National Institute for Health Research Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, United Kingdom.,Oxford University Clinical Research Unit, Ha Noi, Viet Nam
| | | | - Brian Faragher
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Ajit Rayamajhi
- Kanti Children's Hospital, Maharajgunj, Kathmandu, Nepal.,National Academy of Medical Sciences, Kathmandu, Nepal
| | | | | | - Geeta Shakya
- National Public Health Laboratory, Teku, Kathmandu, Nepal
| | - Sutee Yoksan
- Institute of Molecular Biosciences, Mahidol University and Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand
| | - Sameer Dixit
- Center for Molecular Dynamics Nepal, Thapathali, Kathmandu, Nepal
| | | | - Bimal Paudel
- National Public Health Laboratory, Teku, Kathmandu, Nepal
| | | | - Tom Solomon
- Institute of Infection and Global Health and National Institute for Health Research Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, United Kingdom.,Walton Centre NHS Foundation Trust, member of Liverpool Health Partners, Liverpool, United Kingdom
| | - Mike J Griffiths
- Institute of Infection and Global Health and National Institute for Health Research Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, United Kingdom.,Alder Hey Children's NHS Trust, member of Liverpool Health Partners, Liverpool, United Kingdom
| |
Collapse
|
11
|
Kulkarni R, Sapkal GN, Kaushal H, Mourya DT. Japanese Encephalitis: A Brief Review on Indian Perspectives. Open Virol J 2018; 12:121-130. [PMID: 30288200 PMCID: PMC6142657 DOI: 10.2174/1874357901812010121] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 03/23/2018] [Accepted: 05/16/2018] [Indexed: 11/22/2022] Open
Abstract
Introduction: Japanese encephalitis (JE) is recently declared as a notifiable disease in India due to its expanding geographical distribution. The disease notification facilitates effective implementation of preventive measures and case management. Expalantion: JE is a vector-borne disease that can be prevented by vaccine administration. It is caused by Japanese encephalitis virus (JEV), belonging to family Flaviviridae. Amongst the known etiological viral encephalitis agents, it is one of the leading viral agents of acute encephalitis syndrome in many Asian countries where it is identified to cause substantial morbidity and mortality as well as disability. Globally, it is responsible for approximately 68,000 clinical cases every year. Conclusion: In the absence of antivirals, patients are given supportive treatment to relieve and stabilize. Amongst available control strategies; vector control is resource intensive while animal and human vaccination are the most effective tool against the disease. This review highlights recent progress focusing challenges with diagnosis and prophylactic interventions.
Collapse
Affiliation(s)
- Reshma Kulkarni
- ICMR-National Institute of Virology, 20-A, Dr. Ambedkar Road, Pune-411001, India
| | - Gajanan N Sapkal
- ICMR-National Institute of Virology, 20-A, Dr. Ambedkar Road, Pune-411001, India
| | - Himanshu Kaushal
- ICMR-National Institute of Virology, 20-A, Dr. Ambedkar Road, Pune-411001, India
| | - Devendra T Mourya
- ICMR-National Institute of Virology, 20-A, Dr. Ambedkar Road, Pune-411001, India
| |
Collapse
|
12
|
Long-term immunogenicity of an initial booster dose of an inactivated, Vero cell culture-derived Japanese encephalitis vaccine (JE-VC) and the safety and immunogenicity of a second JE-VC booster dose in children previously vaccinated with an inactivated, mouse brain-derived Japanese encephalitis vaccine. Vaccine 2018; 36:1398-1404. [DOI: 10.1016/j.vaccine.2018.01.075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/25/2018] [Accepted: 01/29/2018] [Indexed: 01/23/2023]
|
13
|
Comparison of the adjuvanticity of two adjuvant formulations containing de-O-acylated lipooligosaccharide on Japanese encephalitis vaccine in mice. Arch Pharm Res 2017; 41:219-228. [DOI: 10.1007/s12272-017-0985-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 11/10/2017] [Indexed: 12/17/2022]
|
14
|
Asif A, Manzoor S, Tuz-Zahra F, Saalim M, Ashraf M, Ishtiyaq J, Khalid M. Zika Virus: Immune Evasion Mechanisms, Currently Available Therapeutic Regimens, and Vaccines. Viral Immunol 2017; 30:682-690. [PMID: 29028178 DOI: 10.1089/vim.2017.0046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The sudden emergence of infectious pathogens such as Zika virus (ZIKV) holds global health concerns. Recent dissemination of ZIKV from Pacific to Americas with an upsurge of congenital anomalies and Guillain Barre Syndrome (GBS) in adults has created an alarming situation. High-throughput studies are in progress to understand ZIKV's mode of pathogenesis and mechanism of immune escape, yet the pathogenesis remains obscure. Mainly ZIKV's envelope (E) protein and nonstructural proteins (mainly NS1 and NS5) manipulate host cell to support viral immune escape by modulation of the interferon pathway and complement antagonism. The development of direct therapeutics for ZIKV infection is required to overcome the rapidly evolving viral threat. Currently, the existing strategies for ZIKV treatment are only supportive. Although, there is no prophylactic or therapeutic vaccine presently available, however, recent efforts have brought up ZIKV vaccines into clinical trial phase 1. This review presents the highlights of recent advances in understanding immune evasion strategies adapted by ZIKV and existing therapies against the virus.
Collapse
Affiliation(s)
- Arun Asif
- Atta-ur-Rahman School of Applied Bio-Sciences, National University of Sciences and Technology , Islamabad, Pakistan
| | - Sobia Manzoor
- Atta-ur-Rahman School of Applied Bio-Sciences, National University of Sciences and Technology , Islamabad, Pakistan
| | - Fatima Tuz-Zahra
- Atta-ur-Rahman School of Applied Bio-Sciences, National University of Sciences and Technology , Islamabad, Pakistan
| | - Muhammad Saalim
- Atta-ur-Rahman School of Applied Bio-Sciences, National University of Sciences and Technology , Islamabad, Pakistan
| | - Maliha Ashraf
- Atta-ur-Rahman School of Applied Bio-Sciences, National University of Sciences and Technology , Islamabad, Pakistan
| | - Javeria Ishtiyaq
- Atta-ur-Rahman School of Applied Bio-Sciences, National University of Sciences and Technology , Islamabad, Pakistan
| | - Madiha Khalid
- Atta-ur-Rahman School of Applied Bio-Sciences, National University of Sciences and Technology , Islamabad, Pakistan
| |
Collapse
|
15
|
Hegde NR, Gore MM. Japanese encephalitis vaccines: Immunogenicity, protective efficacy, effectiveness, and impact on the burden of disease. Hum Vaccin Immunother 2017; 13:1-18. [PMID: 28301270 DOI: 10.1080/21645515.2017.1285472] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Japanese encephalitis (JE) is a serious public health concern in most of Asia. The disease is caused by JE virus (JEV), a flavivirus transmitted by Culex mosquitoes. Several vaccines have been developed to control JE in endemic areas as well as to protect travelers and military personnel who visit or are commissioned from non-endemic to endemic areas. The vaccines include inactivated vaccines produced in mouse brain or cell cultures, live attenuated vaccines, and a chimeric vaccine based on the live attenuated yellow fever virus 17D vaccine strain. All the marketed vaccines belong to the JEV genotype III, but have been shown to be efficacious against other genotypes and strains, with varying degrees of cross-neutralization, albeit at levels deemed to be protective. The protective responses have been shown to last three or more years, depending on the type of vaccine and the number of doses. This review presents a brief account of the different JE vaccines, their immunogenicity and protective ability, and the impact of JE vaccines in reducing the burden of disease in endemic countries.
Collapse
Affiliation(s)
- Nagendra R Hegde
- a Ella Foundation, Genome Valley , Turkapally, Shameerpet Mandal , Hyderabad , India
| | - Milind M Gore
- b National Institute of Virology, Indian Council of Medical Research , Pune , India
| |
Collapse
|
16
|
Turtle L, Tatullo F, Bali T, Ravi V, Soni M, Chan S, Chib S, Venkataswamy MM, Fadnis P, Yaïch M, Fernandez S, Klenerman P, Satchidanandam V, Solomon T. Cellular Immune Responses to Live Attenuated Japanese Encephalitis (JE) Vaccine SA14-14-2 in Adults in a JE/Dengue Co-Endemic Area. PLoS Negl Trop Dis 2017; 11:e0005263. [PMID: 28135273 PMCID: PMC5279729 DOI: 10.1371/journal.pntd.0005263] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/15/2016] [Indexed: 11/21/2022] Open
Abstract
Background Japanese encephalitis (JE) virus (JEV) causes severe epidemic encephalitis across Asia, for which the live attenuated vaccine SA14-14-2 is being used increasingly. JEV is a flavivirus, and is closely related to dengue virus (DENV), which is co-endemic in many parts of Asia, with clinically relevant interactions. There is no information on the human T cell response to SA14-14-2, or whether responses to SA14-14-2 cross-react with DENV. We used live attenuated JE vaccine SA14-14-2 as a model for studying T cell responses to JEV infection in adults, and to determine whether these T cell responses are cross-reactive with DENV, and other flaviviruses. Methods We conducted a single arm, open label clinical trial (registration: clinicaltrials.gov NCT01656200) to study T cell responses to SA14-14-2 in adults in South India, an area endemic for JE and dengue. Results Ten out of 16 (62.5%) participants seroconverted to JEV SA14-14-2, and geometric mean neutralising antibody (NAb) titre was 18.5. Proliferation responses were commonly present before vaccination in the absence of NAb, indicating a likely high degree of previous flavivirus exposure. Thirteen of 15 (87%) participants made T cell interferon-gamma (IFNγ) responses against JEV proteins. In four subjects tested, at least some T cell epitopes mapped cross-reacted with DENV and other flaviviruses. Conclusions JEV SA14-14-2 was more immunogenic for T cell IFNγ than for NAb in adults in this JE/DENV co-endemic area. The proliferation positive, NAb negative combination may represent a new marker of long term immunity/exposure to JE. T cell responses can cross-react between JE vaccine and DENV in a co-endemic area, illustrating a need for greater knowledge on such responses to inform the development of next-generation vaccines effective against both diseases. Trial Registration clinicaltrials.gov (NCT01656200) The Flavivirus genus member Japanese encephalitis (JE) virus (JEV), causes severe brain disease in tens of thousands of children across Asia every year. JE is vaccine preventable, and the immune response to JEV plays a major role in disease outcome. However, the response to JEV is hard to study as JE affects young children in rural areas. Related flaviviruses, such as dengue virus (which has no good vaccine), can influence the outcome of JE, probably due to cross-reactive immune responses. T cells (a subset of white blood cells) respond to virus infections, but we know little about the timing and nature of T cell responses to JEV after infection and whether T cells are protective against JEV. We used the live JE vaccine SA14-14-2 as a model to study the immune response to JEV. We found T cell responses frequently after JE vaccination. In this small group of volunteers, many of whom were exposed to dengue virus, most of the T cell responses tested cross-reacted between JEV and dengue virus. However, only about two thirds of people made antibody responses to the vaccine. Studying these responses could help design new vaccines for use against JE and dengue in Asia.
Collapse
Affiliation(s)
- Lance Turtle
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
- NIHR Health Protection Research Unit for Emerging and Zoonotic Infections, University of Liverpool, Liverpool, United Kingdom
- Tropical & Infectious Disease Unit, Royal Liverpool University Hospital, Liverpool, United Kingdom
- * E-mail: (LT); (VS)
| | - Filippo Tatullo
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
- Dept of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Tanushka Bali
- Dept of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Vasanthapuram Ravi
- Dept of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Mohammed Soni
- Dept of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Sajesh Chan
- Dept of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Savita Chib
- Dept of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Manjunatha M. Venkataswamy
- Dept of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Prachi Fadnis
- Dept of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | | | - Stefan Fernandez
- Dept of Virology, Armed Forces Research Institute of Medical Science (AFRIMS), Bangkok, Thailand
| | - Paul Klenerman
- Translational Gastroenterology Unit, Nuffield Dept. of Medicine, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, United Kingdom
| | - Vijaya Satchidanandam
- Dept of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
- * E-mail: (LT); (VS)
| | - Tom Solomon
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
- NIHR Health Protection Research Unit for Emerging and Zoonotic Infections, University of Liverpool, Liverpool, United Kingdom
- Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
17
|
Sero-Molecular Epidemiology of Japanese Encephalitis in Zhejiang, an Eastern Province of China. PLoS Negl Trop Dis 2016; 10:e0004936. [PMID: 27560360 PMCID: PMC4999095 DOI: 10.1371/journal.pntd.0004936] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/01/2016] [Indexed: 11/19/2022] Open
Abstract
Background Sporadic Japanese encephalitis (JE) cases still have been reported in Zhejiang Province in recent years, and concerns about vaccine cross-protection and population-level immunity have been raised off and on within the public health sphere. Genotype I (GI) has replaced GIII as the dominant genotype in Asian countries during the past few decades, which caused considerable concerns about the potential change of epidemiology characteristics and the vaccine effectiveness. The aim of this study was to investigate the prevalence of JE neutralizing antibody and its waning antibody trend after live attenuated JE vaccine immunization. Additionally, this study analyzed the molecular characteristics of the E gene of Zhejiang Japanese encephalitis virus (JEV) strains, and established genetic relationships with other JEV strains. Methodology/Principal Findings A total of 570 serum specimens were sampled from community population aged from 0 to 92 years old in Xianju county of Zhejiang Province in 2013–2014. Microseroneutralization test results were analyzed to estimate the population immunity and to observe antibody dynamics in vaccinated children. E genes of 28 JEV strains isolated in Zhejiang Province were sequenced for phylogenetic tree construction and molecular characteristics analysis with other selected strains. Positive JE neutralizing antibody rates were higher in residents ≥35 years old (81%~98%) and lower in residents <35 years old (0~57%). 7 or 8 years after the 2nd live attenuated vaccine dose, the antibodies against for 4 different strains with microseroneutralization test were decreased by 55%~73% on seropositive rates and by 25%~38% on GMTs respectively. JEV strains isolated in recent years were all grouped into GI, while those isolated in the 1980s belonged to GIII. On important amino acid sites related to antigenicity, there was no divergence between the Zhejiang JE virus strains and the vaccine strain (SA14-14-2). Conclusion/Significances JE neutralizing antibody positive rates increase in age ≥10 years old population, likely reflecting natural infection or natural boosting of immunity through exposure to wild virus. JE seropositivity rates were quite low in <35 years old age groups in Zhejiang Province. Waning of neutralizing antibody after live attenuated vaccine immunization was observed, but the clinical significance should be further investigated. Both the peripheral antibody response and genetic characterization indicate that current live attenuated JE vaccine conferred equal neutralizing potency against GI or GIII of wild strains. GI has replaced GIII as the dominant genotype in Zhejiang in the past few decades. Although the chance of exposure to wild JE virus has reduced, the virus still circulates in nature; therefore, it is necessary to implement immunization program for children continually and to conduct surveillance activity periodically. Japanese encephalitis (JE) remains one of the most significant public health problems in Asia and the Western Pacific region. A JE viral infection can cause death and severe sequelae. Vaccination is the most effective method for preventing JE currently. After decades of routine vaccination, the number of JE cases declined considerably in Zhejiang Province, China. However, emergence of genotype I of JE as the most common genotype in China in recent decades has become a major public health problem. As all the currently available vaccines are derived from genotype III strains, the circulations of another genotype have caused considerable concerns about vaccine effectiveness. In this study, we found that population immunity against JE was quite low in children and adolescents. Waning of JE neutralizing antibody after JE immunization was observed. Therefore, issues about duration of protection and booster dose necessity need further research. On the bright side, evidence shows that the JE vaccine currently used is effective for both genotype I and III of wild viruses. Although clinical JE cases have reduced, the virus is still spreading in nature; therefore, we encourage children and other high-risk groups to adhere to the immunization program continuously.
Collapse
|
18
|
Immunogenicity and Safety of a Booster Dose of a Live Attenuated Japanese Encephalitis Chimeric Vaccine Given 1 Year After Primary Immunization in Healthy Children in the Republic of Korea. Pediatr Infect Dis J 2016; 35:e60-4. [PMID: 26535878 DOI: 10.1097/inf.0000000000000967] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND This study evaluated the effect of a booster vaccination of a new, live attenuated, Japanese encephalitis chimeric vaccine (JE-CV). Previously this vaccine has been used as a booster 12 months after priming with an inactivated vaccine and at >24 months after priming with the same JE-CV. This study evaluates the immunogenicity and safety of the JE-CV given at 12-24 months after JE-CV priming. METHODS Phase III, open-label study in the Republic of Korea in which 119 children previously vaccinated with JE-CV at 12-24 months of age received a JE-CV booster at 12-24 months after primary vaccination. JE neutralizing antibody titers were measured using >50% plaque reduction neutralization test prebooster and 1 month postbooster vaccination. Seroprotection (SP) was defined as ≥10 (1/dil). Safety was assessed for 28 days postvaccination by parental reports. Serious adverse events were monitored for 6 months postvaccination. RESULTS Antibody persistence was high prebooster (SP rate 93.5%). There was a strong anamnestic response postbooster vaccination, with an SP rate of 100% and a >50-fold increase in geometric mean titer from the prebooster level. Both antibody persistence and the booster response were independent of whether the booster was given at 12-17 or 18-24 months. The safety profile was good and comparable with the primary vaccination; there were no vaccine-related serious adverse events and no deaths. CONCLUSIONS This study confirms the suitability of a JE-CV booster vaccination at 12-24 months after a primary dose of the same vaccine given at 12-24 months of age in children in the Republic of Korea.
Collapse
|
19
|
Jamrozik E, Selgelid MJ. Ethics, Climate Change and Infectious Disease. BIOETHICAL INSIGHTS INTO VALUES AND POLICY 2016. [DOI: 10.1007/978-3-319-26167-6_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
20
|
Nah JJ, Yang DK, Kim HH, Song JY. The present and future of veterinary vaccines for Japanese encephalitis in Korea. Clin Exp Vaccine Res 2015; 4:130-6. [PMID: 26273571 PMCID: PMC4524897 DOI: 10.7774/cevr.2015.4.2.130] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/03/2015] [Accepted: 05/20/2015] [Indexed: 12/30/2022] Open
Abstract
Japanese encephalitis (JE) is a mosquito-borne zoonotic disease that affects approximately 50,000 people annually in Asia, causing 10,000 deaths. Considering the role of pigs as the virus-amplifying host and the economic loss in the swine industry, JE is an important disease for both public and animal health. A nationwide JE virus (JEV) vaccination program has been conducted annually for more than 30 years to prevent severe reproductive disorders in the Korean sow population. Remarkable progress in molecular biology has made it possible to analyze the genome of the vaccine strain at the nucleotide and amino acid levels. However, the scientific record of the current JEV veterinary vaccine has not been reported. Therefore, this article outlines the current JEV vaccine strain used in animals and discusses future directions for developing new veterinary JEV vaccines.
Collapse
Affiliation(s)
- Jin-Ju Nah
- Animal and Plant Quarantine Agency, MAFRA, Anyang, Korea
| | - Dong-Kun Yang
- Animal and Plant Quarantine Agency, MAFRA, Anyang, Korea
| | - Ha-Hyun Kim
- Animal and Plant Quarantine Agency, MAFRA, Anyang, Korea
| | - Jae-Young Song
- Animal and Plant Quarantine Agency, MAFRA, Anyang, Korea
| |
Collapse
|
21
|
Kim JK, Kim JM, Song BH, Yun SI, Yun GN, Byun SJ, Lee YM. Profiling of viral proteins expressed from the genomic RNA of Japanese encephalitis virus using a panel of 15 region-specific polyclonal rabbit antisera: implications for viral gene expression. PLoS One 2015; 10:e0124318. [PMID: 25915765 PMCID: PMC4410938 DOI: 10.1371/journal.pone.0124318] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 02/27/2015] [Indexed: 12/16/2022] Open
Abstract
Japanese encephalitis virus (JEV), a mosquito-borne flavivirus, is closely related to West Nile (WN), yellow fever (YF), and dengue (DEN) viruses. Its plus-strand genomic RNA carries a single open reading frame encoding a polyprotein that is cleaved into three structural (C, prM/M, and E) and at least seven nonstructural (NS1/NS1', NS2A, NS2B, NS3, NS4A, NS4B, and NS5) proteins, based on previous work with WNV, YFV, and DENV. Here, we aimed to profile experimentally all the viral proteins found in JEV-infected cells. We generated a collection of 15 JEV-specific polyclonal antisera covering all parts of the viral protein-coding regions, by immunizing rabbits with 14 bacterially expressed glutathione-S-transferase fusion proteins (for all nine viral proteins except NS2B) or with a chemically synthesized oligopeptide (for NS2B). In total lysates of JEV-infected BHK-21 cells, immunoblotting with these antisera revealed: (i) three mature structural proteins (~12-kDa C, ~8-kDa M, and ~53-kDa E), a precursor of M (~24-kDa prM) and three other M-related proteins (~10-14 kDa); (ii) the predicted ~45-kDa NS1 and its frameshift product, ~58-kDa NS1', with no evidence of the predicted ~25-kDa NS2A; (iii) the predicted but hardly detectable ~14-kDa NS2B and an unexpected but predominant ~12-kDa NS2B-related protein; (iv) the predicted ~69-kDa NS3 plus two major cleavage products (~34-kDa NS3N-term and ~35-kDa NS3C-term), together with at least nine minor proteins of ~16-52 kDa; (v) the predicted ~14-kDa NS4A; (vi) two NS4B-related proteins (~27-kDa NS4B and ~25-kDa NS4B'); and (vii) the predicted ~103-kDa NS5 plus at least three other NS5-related proteins (~15 kDa, ~27 kDa, and ~90 kDa). Combining these data with confocal microscopic imaging of the proteins' intracellular localization, our study is the first to provide a solid foundation for the study of JEV gene expression, which is crucial for elucidating the regulatory mechanisms of JEV genome replication and pathobiology.
Collapse
Affiliation(s)
- Jin-Kyoung Kim
- Department of Animal, Dairy, and Veterinary Sciences; Utah Science Technology and Research, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
| | - Jeong-Min Kim
- Department of Microbiology, College of Medicine, Chungbuk National University, Cheongju, South Korea
| | - Byung-Hak Song
- Department of Animal, Dairy, and Veterinary Sciences; Utah Science Technology and Research, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
| | - Sang-Im Yun
- Department of Animal, Dairy, and Veterinary Sciences; Utah Science Technology and Research, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
| | - Gil-Nam Yun
- Department of Microbiology, College of Medicine, Chungbuk National University, Cheongju, South Korea
| | - Sung-June Byun
- Animal Biotechnology Division, Korea National Institute of Animal Science, Suwon, South Korea
| | - Young-Min Lee
- Department of Animal, Dairy, and Veterinary Sciences; Utah Science Technology and Research, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
- * E-mail:
| |
Collapse
|
22
|
Ye Q, Xu YP, Zhang Y, Li XF, Wang HJ, Liu ZY, Li SH, Liu L, Zhao H, Nian QG, Deng YQ, Qin ED, Qin CF. Genotype-specific neutralization determinants in envelope protein: implications for the improvement of Japanese encephalitis vaccine. J Gen Virol 2015; 96:2165-2175. [PMID: 25908779 DOI: 10.1099/vir.0.000160] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Japanese encephalitis remains the leading cause of viral encephalitis in children in Asia and is expanding its geographical range to larger areas in Asia and Australasia. Five genotypes of Japanese encephalitis virus (JEV) co-circulate in the geographically affected areas. In particular, the emergence of genotype I (GI) JEV has displaced genotype III (GIII) as the dominant circulating genotype in many Asian regions. However, all approved vaccine products are derived from GIII strains. In the present study, bioinformatic analysis revealed that GI and GIII JEV strains shared two distinct amino acid residues within the envelope (E) protein (E222 and E327). By using reverse genetics approaches, A222S and S327T mutations were demonstrated to decrease live-attenuated vaccine (LAV) SA14-14-2-induced neutralizing antibodies in humans, without altering viral replication. A222S or S327T mutations were then rationally engineered into the infectious clone of SA14-14-2, and the resulting mutant strains retained the same genetic stability and attenuation characteristics as the parent strain. More importantly, immunization of mice with LAV-A222S or LAV-S327T elicited increased neutralizing antibodies against GI strains. Together, these results demonstrated that E222 and E327 are potential genotype-related neutralization determinants and are critical in determining the protective efficacy of live Japanese encephalitis vaccine SA14-14-2 against circulating GI strains. Our findings will aid in the rational design of the next generation of Japanese encephalitis LAVs capable of providing broad protection against all JEV strains belonging to different genotypes.
Collapse
Affiliation(s)
- Qing Ye
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China
| | - Yan-Peng Xu
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China
| | - Yu Zhang
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China
| | - Xiao-Feng Li
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China.,State Key Laboratory of Pathogen and Biosecurity, Beijing 100071, PR China
| | - Hong-Jiang Wang
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China
| | - Zhong-Yu Liu
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China
| | - Shi-Hua Li
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China
| | - Long Liu
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China.,Graduate School, Anhui Medical University, Hefei 230032, PR China
| | - Hui Zhao
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China.,State Key Laboratory of Pathogen and Biosecurity, Beijing 100071, PR China
| | - Qing-Gong Nian
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China
| | - Yong-Qiang Deng
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China
| | - E-De Qin
- Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China
| | - Cheng-Feng Qin
- Graduate School, Anhui Medical University, Hefei 230032, PR China.,Department of Virology, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China.,State Key Laboratory of Pathogen and Biosecurity, Beijing 100071, PR China
| |
Collapse
|
23
|
Genetic Determinants of Japanese Encephalitis Virus Vaccine Strain SA14-14-2 That Govern Attenuation of Virulence in Mice. J Virol 2015; 89:6328-37. [PMID: 25855730 DOI: 10.1128/jvi.00219-15] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/30/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The safety and efficacy of the live-attenuated Japanese encephalitis virus (JEV) SA14-14-2 vaccine are attributed to mutations that accumulated in the viral genome during its derivation. However, little is known about the contribution that is made by most of these mutations to virulence attenuation and vaccine immunogenicity. Here, we generated recombinant JEV (rJEV) strains containing JEV SA14-14-2 vaccine-specific mutations that are located in the untranslated regions (UTRs) and seven protein genes or are introduced from PCR-amplified regions of the JEV SA14-14-2 genome. The resulting mutant viruses were evaluated in tissue culture and in mice. The authentic JEV SA14-14-2 (E) protein, with amino acid substitutions L107F, E138K, I176V, T177A, E244G, Q264H, K279M, A315V, S366A, and K439R relative to the wild-type rJEV clone, was essential and sufficient for complete attenuation of neurovirulence. Individually, the nucleotide substitution T39A in the 5' UTR (5'-UTR-T39A), the capsid (C) protein amino acid substitution L66S (C-L66S), and the complete NS1/2A genome region containing 10 mutations each significantly reduced virus neuroinvasion but not neurovirulence. The levels of peripheral virulence attenuation imposed by the 5'-UTR-T39A and C-L66S mutations, individually, were somewhat mitigated in combination with other vaccine strain-specific mutations, which might be compensatory, and together did not affect immunogenicity. However, a marked reduction in immunogenicity was observed with the addition of the NS1/2A and NS5 vaccine virus genome regions. These results suggest that a second-generation recombinant vaccine can be rationally engineered to maximize levels of immunogenicity without compromising safety. IMPORTANCE The live-attenuated JEV SA14-14-2 vaccine has been vital for controlling the incidence of disease caused by JEV, particularly in rural areas of Asia where it is endemic. The vaccine was developed >25 years ago by passaging wild-type JEV strain SA14 in tissue cultures and rodents, with intermittent tissue culture plaque purifications, to produce a virus clone that had adequate levels of attenuation and immunogenicity. The vaccine and parent virus sequences were later compared, and mutations were identified throughout the vaccine virus genome, but their contributions to attenuation were never fully elucidated. Here, using reverse genetics, we comprehensively defined the impact of JEV SA14-14-2 mutations on attenuation of virulence and immunogenicity in mice. These results are relevant for quality control of new lots of the current live-attenuated vaccine and provide insight for the rational design of second-generation, live-attenuated, recombinant JEV vaccine candidates.
Collapse
|
24
|
Safety and immunogenicity of a freeze-dried, Vero cell culture-derived, inactivated Japanese encephalitis vaccine (KD-287, ENCEVAC®) versus a mouse brain-derived inactivated Japanese encephalitis vaccine in children: a phase III, multicenter, double-blinded, randomized trial. BMC Infect Dis 2015; 15:7. [PMID: 25567119 PMCID: PMC4296691 DOI: 10.1186/s12879-014-0744-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 12/24/2014] [Indexed: 11/10/2022] Open
Abstract
Background Although mouse brain-derived, inactivated Japanese encephalitis vaccines (JE-MBs) have been successfully used for a long time, potential rare neurological complications have prompted the development of a Vero cell culture-derived inactivated vaccine (JE-VC). In a phase III clinical study, we aimed to compare the safety and immunogenicity of a JE-VC, KD-287 with a JE-MB, JEV-GCC, in children. Methods In this multicenter, double-blinded, randomized controlled trial, the study population consisted of 205 healthy Korean children aged 12–23 months. Each subject was subcutaneously vaccinated with either KD-287 or JEV-GCC twice at an interval of 2 weeks and then vaccinated once 12 months after the second vaccination. Neutralizing antibodies were measured by the plaque reduction neutralization test using the homologous and heterologous, as a post hoc analysis, challenge virus strains. Results The three-dose regimen of KD-287 showed a comparable safety profile with JEV-GCC except higher incidence of fever after the first dose (30.4% and 14.7%, respectively). Most of the fever was mild degree (61.3% and 66.7%, respectively). KD-287 fulfilled the non-inferiority criteria for seroconversion rate (SCR) and geometric mean titer (GMT) of the neutralizing antibody, which were the primary endpoints, at 4 weeks after the third vaccination (95% CI: −1.00, 3.10 for the SCR difference and 10.8, 17.6 for the GMT ratio). The SCRs of KD-287 were all 100% and the GMTs were higher in the KD-287 group than in the JEV-GCC group after the second vaccination and before and after the third vaccination (GMT ratio: 5.59, 20.13, and 13.79, respectively, p < 0.001 in all). GMTs were higher in the KD-287 group in the heterologous analysis also (GMT ratio: 4.05, 5.15, and 4.19, respectively, p < 0.001 in all). Conclusions This study suggests that the KD-287, a JE-VC is as safe as and may be more effective than the licensed MB-derived vaccine. KD-287 could thus be useful as a second-generation vaccine and substitute for the current JE-MB vaccine in Korean children. Trial registration ClinicalTrials.gov: NCT01150942 Electronic supplementary material The online version of this article (doi:10.1186/s12879-014-0744-4) contains supplementary material, which is available to authorized users.
Collapse
|
25
|
Thomas SJ, Endy TP, Rothman AL, Barrett AD. Flaviviruses (Dengue, Yellow Fever, Japanese Encephalitis, West Nile Encephalitis, St. Louis Encephalitis, Tick-Borne Encephalitis, Kyasanur Forest Disease, Alkhurma Hemorrhagic Fever, Zika). MANDELL, DOUGLAS, AND BENNETT'S PRINCIPLES AND PRACTICE OF INFECTIOUS DISEASES 2015:1881-1903.e6. [DOI: 10.1016/b978-1-4557-4801-3.00155-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
26
|
Abstract
In this chapter, we describe 73 zoonotic viruses that were isolated in Northern Eurasia and that belong to the different families of viruses with a single-stranded RNA (ssRNA) genome. The family includes viruses with a segmented negative-sense ssRNA genome (families Bunyaviridae and Orthomyxoviridae) and viruses with a positive-sense ssRNA genome (families Togaviridae and Flaviviridae). Among them are viruses associated with sporadic cases or outbreaks of human disease, such as hemorrhagic fever with renal syndrome (viruses of the genus Hantavirus), Crimean–Congo hemorrhagic fever (CCHFV, Nairovirus), California encephalitis (INKV, TAHV, and KHATV; Orthobunyavirus), sandfly fever (SFCV and SFNV, Phlebovirus), Tick-borne encephalitis (TBEV, Flavivirus), Omsk hemorrhagic fever (OHFV, Flavivirus), West Nile fever (WNV, Flavivirus), Sindbis fever (SINV, Alphavirus) Chikungunya fever (CHIKV, Alphavirus) and others. Other viruses described in the chapter can cause epizootics in wild or domestic animals: Geta virus (GETV, Alphavirus), Influenza A virus (Influenzavirus A), Bhanja virus (BHAV, Phlebovirus) and more. The chapter also discusses both ecological peculiarities that promote the circulation of these viruses in natural foci and factors influencing the occurrence of epidemic and epizootic outbreaks
Collapse
|
27
|
Gromowski GD, Firestone CY, Bustos-Arriaga J, Whitehead SS. Genetic and phenotypic properties of vero cell-adapted Japanese encephalitis virus SA14-14-2 vaccine strain variants and a recombinant clone, which demonstrates attenuation and immunogenicity in mice. Am J Trop Med Hyg 2014; 92:98-107. [PMID: 25311701 DOI: 10.4269/ajtmh.14-0427] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The live-attenuated Japanese encephalitis virus (JEV) SA14-14-2 vaccine, produced in primary hamster kidney cells, is safe and effective. Past attempts to adapt this virus to replicate in cells that are more favorable for vaccine production resulted in mutations that significantly reduced immunogenicity. In this study, 10 genetically distinct Vero cell-adapted JEV SA14-14-2 variants were isolated and a recombinant wild-type JEV clone, modified to contain the JEV SA14-14-2 polyprotein amino acid sequence, was recovered in Vero cells. A single capsid protein mutation (S66L) was important for Vero cell-adaptation. Mutations were also identified that modulated virus sensitivity to type I interferon-stimulation in Vero cells. A subset of JEV SA14-14-2 variants and the recombinant clone were evaluated in vivo and exhibited levels of attenuation that varied significantly in suckling mice, but were avirulent and highly immunogenic in weanling mice and are promising candidates for the development of a second-generation, recombinant vaccine.
Collapse
Affiliation(s)
- Gregory D Gromowski
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Cai-Yen Firestone
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - José Bustos-Arriaga
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Stephen S Whitehead
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
28
|
Yun SI, Song BH, Kim JK, Yun GN, Lee EY, Li L, Kuhn RJ, Rossmann MG, Morrey JD, Lee YM. A molecularly cloned, live-attenuated japanese encephalitis vaccine SA14-14-2 virus: a conserved single amino acid in the ij Hairpin of the Viral E glycoprotein determines neurovirulence in mice. PLoS Pathog 2014; 10:e1004290. [PMID: 25077483 PMCID: PMC4117607 DOI: 10.1371/journal.ppat.1004290] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 06/18/2014] [Indexed: 01/12/2023] Open
Abstract
Japanese encephalitis virus (JEV), a mosquito-borne flavivirus that causes fatal neurological disease in humans, is one of the most important emerging pathogens of public health significance. JEV represents the JE serogroup, which also includes West Nile, Murray Valley encephalitis, and St. Louis encephalitis viruses. Within this serogroup, JEV is a vaccine-preventable pathogen, but the molecular basis of its neurovirulence remains unknown. Here, we constructed an infectious cDNA of the most widely used live-attenuated JE vaccine, SA14-14-2, and rescued from the cDNA a molecularly cloned virus, SA14-14-2MCV, which displayed in vitro growth properties and in vivo attenuation phenotypes identical to those of its parent, SA14-14-2. To elucidate the molecular mechanism of neurovirulence, we selected three independent, highly neurovirulent variants (LD50, <1.5 PFU) from SA14-14-2MCV (LD50, >1.5×105 PFU) by serial intracerebral passage in mice. Complete genome sequence comparison revealed a total of eight point mutations, with a common single G1708→A substitution replacing a Gly with Glu at position 244 of the viral E glycoprotein. Using our infectious SA14-14-2 cDNA technology, we showed that this single Gly-to-Glu change at E-244 is sufficient to confer lethal neurovirulence in mice, including rapid development of viral spread and tissue inflammation in the central nervous system. Comprehensive site-directed mutagenesis of E-244, coupled with homology-based structure modeling, demonstrated a novel essential regulatory role in JEV neurovirulence for E-244, within the ij hairpin of the E dimerization domain. In both mouse and human neuronal cells, we further showed that the E-244 mutation altered JEV infectivity in vitro, in direct correlation with the level of neurovirulence in vivo, but had no significant impact on viral RNA replication. Our results provide a crucial step toward developing novel therapeutic and preventive strategies against JEV and possibly other encephalitic flaviviruses.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Blotting, Northern
- Blotting, Western
- Cloning, Molecular
- Encephalitis Virus, Japanese/genetics
- Encephalitis Virus, Japanese/immunology
- Encephalitis, Japanese/genetics
- Encephalitis, Japanese/immunology
- Encephalitis, Japanese/virology
- Female
- Flow Cytometry
- Humans
- Immunoenzyme Techniques
- Japanese Encephalitis Vaccines/genetics
- Japanese Encephalitis Vaccines/immunology
- Membrane Glycoproteins/chemistry
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred ICR
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Mutation/genetics
- Nervous System/virology
- Protein Conformation
- Sequence Homology, Amino Acid
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Viral Envelope Proteins/chemistry
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/metabolism
- Virulence/genetics
- Virus Replication
Collapse
Affiliation(s)
- Sang-Im Yun
- Department of Animal, Dairy, and Veterinary Sciences; Utah Science Technology and Research, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
| | - Byung-Hak Song
- Department of Animal, Dairy, and Veterinary Sciences; Utah Science Technology and Research, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
| | - Jin-Kyoung Kim
- Department of Animal, Dairy, and Veterinary Sciences; Utah Science Technology and Research, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
| | - Gil-Nam Yun
- Department of Microbiology, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Eun-Young Lee
- Department of Anatomy, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Long Li
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Richard J. Kuhn
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Michael G. Rossmann
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - John D. Morrey
- Department of Animal, Dairy, and Veterinary Sciences; Utah Science Technology and Research, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
| | - Young-Min Lee
- Department of Animal, Dairy, and Veterinary Sciences; Utah Science Technology and Research, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
| |
Collapse
|
29
|
Gromowski GD, Firestone CY, Hanson CT, Whitehead SS. Japanese encephalitis virus vaccine candidates generated by chimerization with dengue virus type 4. Vaccine 2014; 32:3010-8. [PMID: 24699473 DOI: 10.1016/j.vaccine.2014.03.062] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 03/10/2014] [Accepted: 03/17/2014] [Indexed: 10/25/2022]
Abstract
Japanese encephalitis virus (JEV) is a leading cause of viral encephalitis worldwide and vaccination is one of the most effective ways to prevent disease. A suitable live-attenuated JEV vaccine could be formulated with a live-attenuated tetravalent dengue vaccine for the control of these viruses in endemic areas. Toward this goal, we generated chimeric virus vaccine candidates by replacing the precursor membrane (prM) and envelope (E) protein structural genes of recombinant dengue virus type 4 (rDEN4) or attenuated vaccine candidate rDEN4Δ30 with those of wild-type JEV strain India/78. Mutations were engineered in E, NS3 and NS4B protein genes to improve replication in Vero cells. The chimeric viruses were attenuated in mice and some elicited modest but protective levels of immunity after a single dose. One particular chimeric virus, bearing E protein mutation Q264H, replicated to higher titer in tissue culture and was significantly more immunogenic in mice. The results are compared with live-attenuated JEV vaccine strain SA14-14-2.
Collapse
Affiliation(s)
- Gregory D Gromowski
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.
| | - Cai-Yen Firestone
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Christopher T Hanson
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Stephen S Whitehead
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
30
|
Bonaparte M, Dweik B, Feroldi E, Meric C, Bouckenooghe A, Hildreth S, Hu B, Yoksan S, Boaz M. Immune response to live-attenuated Japanese encephalitis vaccine (JE-CV) neutralizes Japanese encephalitis virus isolates from south-east Asia and India. BMC Infect Dis 2014; 14:156. [PMID: 24656175 PMCID: PMC3994458 DOI: 10.1186/1471-2334-14-156] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 03/14/2014] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND During clinical development of the licensed Japanese encephalitis chimeric virus vaccine (JE-CV), the neutralization capacity of vaccine-induced antibodies was assessed against the vaccine virus and against well characterized wild-type (wt) viruses isolated between 1949-1991. We assessed whether JE-CV-induced antibodies can also neutralize more recent wt Japanese encephalitis virus (JEV) isolates including a genotype 1 isolate. METHODS Sera from 12-18 month-old children who received a single dose of JE-CV in a phase III study in Thailand and the Philippines (ClinicalTrials.gov NCT00735644) were randomly selected and pooled according to neutralization titer against JE-CV into eight samples. Neutralization was assessed by plaque reduction neutralization tests (PRNT50) against three recent isolates from JEV genotypes 1 and 3 in addition to four JEV previously tested. RESULTS Neutralization titers against the three recent JEV strains were comparable to those observed previously against other strains and the vaccine virus. The observed differences between responses to genotype 1 and 3 viruses were within assay variability for the PRNT50. CONCLUSIONS The results were consistent with previously generated data on the neutralization of wt JEV isolates, immune responses induced by JE-CV neutralize recently isolated virus from southeast (SE) Asia and India.
Collapse
Affiliation(s)
- Matthew Bonaparte
- Sanofi Pasteur Global Clinical Immunology Department, Swiftwater, USA
| | - Bashir Dweik
- Sanofi Pasteur Global Clinical Immunology Department, Swiftwater, USA
| | - Emmanuel Feroldi
- Sanofi Pasteur Clinical Development Department, Marcy l’Etoile, France
| | - Claude Meric
- Sanofi Pasteur Clinical Development Department, Marcy l’Etoile, France
| | | | - Stephen Hildreth
- Sanofi Pasteur Global Clinical Immunology Department, Swiftwater, USA
| | - Branda Hu
- Sanofi Pasteur Global Clinical Immunology Department, Swiftwater, USA
| | - Sutee Yoksan
- Center for Vaccine Development, Institute of Molecular Bioscience, Mahidol University, Bangkok, Thailand
| | - Mark Boaz
- Sanofi Pasteur Global Clinical Immunology Department, Swiftwater, USA
| |
Collapse
|
31
|
Concomitant administration of live attenuated Japanese encephalitis chimeric virus vaccine (JE-CV) and measles, mumps, rubella (MMR) vaccine: randomized study in toddlers in Taiwan. Vaccine 2014; 32:5363-9. [PMID: 24631095 DOI: 10.1016/j.vaccine.2014.02.085] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 02/18/2014] [Accepted: 02/26/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Japanese encephalitis (JE) is the most important cause of viral encephalitis in Asia. METHODS In this randomized, open-label, multicenter trial in 550 children aged 12 to 18 months in Taiwan, children received one dose of JE-CV and one dose of MMR vaccine. Vaccines were either administered separately 6 weeks apart (Groups 'JE-CV' and 'MMR', named after which vaccine was given first), or concomitantly (Group 'Co-Ad'). JE neutralizing antibody titers were assessed using PRNT50. MMR antibody levels were determined by ELISA. RESULTS All groups had low seroprotection/seropositivity rates (<10%) before vaccination for all antigens. Forty two days after vaccination, on either Study Day 42 or 84, seroconversion rates for all antigens were high in all groups, irrespective of the order of vaccinations. Seroconversion for JE ranged from 96.9% in Group Co-Ad on D42 to 100% in Group MMR. Non-inferiority was demonstrated for all analyses as the lower bound of the 95% CI of the difference in seroconversion rates between groups was above the pre-defined limit of -10.0%. The immune responses remained high for all antigens and well above the level of protection 12 months after vaccination in all groups. There were no safety concerns. CONCLUSIONS JE-CV is safe and induces a strong protective immune response which persists over 1 year when co-administered with MMR vaccine.
Collapse
|
32
|
A review of successful flavivirus vaccines and the problems with those flaviviruses for which vaccines are not yet available. Vaccine 2014; 32:1326-37. [PMID: 24486372 DOI: 10.1016/j.vaccine.2014.01.040] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 01/11/2014] [Accepted: 01/15/2014] [Indexed: 12/19/2022]
Abstract
Genus flavivirus comprises many important human pathogens causing public health problems worldwide. Some flavivirus infections are characterized by a relatively high mortality rate and/or high sequelae rate in survivors. Because most flavivirus life cycles are maintained between arthropod vectors and amplifying/reservoir hosts in the absence of humans, eradication of flaviviruses might be extremely difficult. Flavivirus vaccine development is considered a reasonable method to prevent flavivirus infections. Some vaccines have been successfully developed, but others have not, regardless of much effort. This review article describes currently available flavivirus vaccines against yellow fever, Japanese encephalitis, and tick-borne encephalitis. In addition, the current status of dengue and West Nile virus vaccine development is reviewed and problems regarding their development are discussed.
Collapse
|
33
|
Halstead SB, Thomas SJ. New Japanese encephalitis vaccines: alternatives to production in mouse brain. Expert Rev Vaccines 2014; 10:355-64. [DOI: 10.1586/erv.11.7] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
34
|
|
35
|
Griffiths MJ, Turtle L, Solomon T. Japanese encephalitis virus infection. HANDBOOK OF CLINICAL NEUROLOGY 2014; 123:561-76. [PMID: 25015504 DOI: 10.1016/b978-0-444-53488-0.00026-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Michael J Griffiths
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK; Alder Hey Children's NHS Foundation Trust, Liverpool, UK.
| | - Lance Turtle
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Tom Solomon
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK; Walton Centre NHS Foundation Trust, Liverpool, UK; NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, UK
| |
Collapse
|
36
|
Development of a small animal peripheral challenge model of Japanese encephalitis virus using interferon deficient AG129 mice and the SA14-14-2 vaccine virus strain. Vaccine 2013; 32:258-64. [PMID: 24252694 DOI: 10.1016/j.vaccine.2013.11.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/28/2013] [Accepted: 11/06/2013] [Indexed: 12/25/2022]
Abstract
Japanese encephalitis virus (JEV) is the most common cause of viral encephalitis in Asia, and it is increasingly a global public health concern due to its recent geographic expansion. While commercial vaccines are available and used in some endemic countries, JEV continues to be a public health problem, with 50,000 cases reported annually. Research with virulent JEV in mouse models to develop new methods of prevention and treatment is restricted to BSL-3 containment facilities, confining these studies to investigators with access to these facilities. We have developed an adult small animal peripheral challenge model using interferon-deficient AG129 mice and the JEV live-attenuated vaccine SA14-14-2, thus requiring only BSL-2 containment. A low dose of virus (10PFU/0.1ml) induced 100% morbidity in infected mice. Increased body temperatures measured by implantable temperature transponders correlated with an increase in infectious virus and viral RNA in serum, spleen and brain as well as an increase in pro-inflammatory markers measured by a 58-biomarker multi-analyte profile (MAP) constructed during the course of infection. In the future, the MAP measurements can be used as a baseline for comparison in order to better assess the inhibition of disease progression by other prophylactic and therapeutic agents. The use of the AG129/JEV SA14-14-2 animal model makes vaccine and therapeutic studies feasible for laboratories with limited biocontainment facilities.
Collapse
|
37
|
Yun SI, Lee YM. Japanese encephalitis: the virus and vaccines. Hum Vaccin Immunother 2013; 10:263-79. [PMID: 24161909 PMCID: PMC4185882 DOI: 10.4161/hv.26902] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 10/14/2013] [Accepted: 10/22/2013] [Indexed: 12/11/2022] Open
Abstract
Japanese encephalitis (JE) is an infectious disease of the central nervous system caused by Japanese encephalitis virus (JEV), a zoonotic mosquito-borne flavivirus. JEV is prevalent in much of Asia and the Western Pacific, with over 4 billion people living at risk of infection. In the absence of antiviral intervention, vaccination is the only strategy to develop long-term sustainable protection against JEV infection. Over the past half-century, a mouse brain-derived inactivated vaccine has been used internationally for active immunization. To date, however, JEV is still a clinically important, emerging, and re-emerging human pathogen of global significance. In recent years, production of the mouse brain-derived vaccine has been discontinued, but 3 new cell culture-derived vaccines are available in various parts of the world. Here we review current aspects of JEV biology, summarize the 4 types of JEV vaccine, and discuss the potential of an infectious JEV cDNA technology for future vaccine development.
Collapse
Affiliation(s)
- Sang-Im Yun
- Department of Animal, Dairy, and Veterinary Sciences; Utah Science Technology and Research; College of Agriculture and Applied Sciences; Utah State University; Logan, UT USA
| | - Young-Min Lee
- Department of Animal, Dairy, and Veterinary Sciences; Utah Science Technology and Research; College of Agriculture and Applied Sciences; Utah State University; Logan, UT USA
| |
Collapse
|
38
|
Kant Upadhyay R. Japanese Encephalitis Virus Generated Neurovirulence, Antigenicity, and Host Immune Responses. ACTA ACUST UNITED AC 2013. [DOI: 10.5402/2013/830396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In response to a JE virus attack, infected body cells start secretion of different cytokines and activate innate immune response. Virus starts neuronal invasion by entering into nerve cells and inflecting the central nervous system. It avoids exposure of body’s natural immunity and generates neurotrophic effects. Virus causes acute susceptibility to CNS and establishes encephalitis syndrome that results in very high fatality in children. In survivors, JEV inhibits the growth and proliferation of NCPs and imposes permanent neuronal disorders like cognitive, motor, and behavioral impairments. However, body cells start TCR mediated interactions, to recognize viral antigens with class I MHC complex on specific target cells, and operate mass killing of virus infected cells by increased CTL activity. Thus, both cell mediated and antibody interactions plays a central role in protection against JEV. In the present review article virus generated neurovirulence, antigenicity, and host immune responses are described in detail. More emphasis is given on diagnosis, clinical care, and active immunization with well-designed potential antiflavivirus vaccines. Further, for achieving an elite success against JEV, global eradication strategies are to be needed for making vaccination program more responsible and effective in endemic areas.
Collapse
Affiliation(s)
- Ravi Kant Upadhyay
- Department of Zoology, D D U Gorakhpur University, Gorakhpur 273009, India
| |
Collapse
|
39
|
Feroldi E, Capeding MR, Boaz M, Gailhardou S, Meric C, Bouckenooghe A. Memory immune response and safety of a booster dose of Japanese encephalitis chimeric virus vaccine (JE-CV) in JE-CV-primed children. Hum Vaccin Immunother 2013; 9:889-97. [PMID: 23442823 DOI: 10.4161/hv.23087] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Japanese encephalitis chimeric virus vaccine (JE-CV) is a licensed vaccine indicated in a single dose administration for primary immunization. This controlled phase III comparative trial enrolled children aged 36-42 mo in the Philippines. 345 children who had received one dose of JE-CV in a study two years earlier, received a JE-CV booster dose. 105 JE-vaccine-naïve children in general good health were randomized to receive JE-CV (JE-vaccine naïve group; 46 children) or varicella vaccine (safety control group; 59 children). JE neutralizing antibody titers were assessed using PRNT50. Immunological memory was observed in children who had received the primary dose of JE-CV before. Seven days after the JE-CV booster dose administration, 96.2% and 66.8% of children were seroprotected and had seroconverted, respectively, and the geometric mean titer (GMT) was 231 1/dil. Twenty-eight days after the JE-CV booster dose seroprotection and seroconversion were achieved in 100% and 95.3% of children, respectively, and the GMT was 2,242 1/dil. In contrast, only 15.4% of JE-CV-vaccine naïve children who had not received any prior JE vaccine were seroprotected seven days after they received JE-CV. One year after receiving the JE-CV booster dose, 99.4% of children remained seroprotected. We conclude that JE-CV is effective and safe, both as a single dose and when administrated as a booster dose. A booster dose increases the peak GMT above the peak level reached after primary immunization and the antibody persistence is maintained at least one year after the JE-CV booster dose administration. Five year follow up is ongoing.
Collapse
Affiliation(s)
- Emmanuel Feroldi
- Sanofi Pasteur Clinical Development Department; Marcy l'Etoile, France
| | | | | | | | | | | |
Collapse
|
40
|
Evaluation of Japanese encephalitis virus polytope DNA vaccine candidate in BALB/c mice. Virus Res 2012; 170:118-25. [DOI: 10.1016/j.virusres.2012.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 09/12/2012] [Accepted: 09/13/2012] [Indexed: 12/26/2022]
|
41
|
Song BH, Yun GN, Kim JK, Yun SI, Lee YM. Biological and genetic properties of SA₁₄-14-2, a live-attenuated Japanese encephalitis vaccine that is currently available for humans. J Microbiol 2012; 50:698-706. [PMID: 22923123 DOI: 10.1007/s12275-012-2336-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 07/13/2012] [Indexed: 11/29/2022]
Abstract
Japanese encephalitis virus (JEV), a mosquito-borne flavivirus, is a major cause of acute encephalitis, a disease of significance for global public health. In the absence of antiviral therapy to treat JEV infection, vaccination is the most effective method of preventing the disease. In JE-endemic areas, the most widely used vaccine to date is SA(14)-14-2, a live-attenuated virus derived from its virulent parent SA(14). In this study, we describe the biological properties of SA(14)-14-2, both in vitro and in vivo, and report the genetic characteristics of its genomic RNA. In BHK-21 (hamster kidney) cells, SA(14)-14-2 displayed a slight delay in plaque formation and growth kinetics when compared to a virulent JEV strain, CNU/LP2, with no decrease in maximum virus production. The delay in viral growth was also observed in two other cell lines, SH-SY5Y (human neuroblastoma) and C6/36 (mosquito larva), which are potentially relevant to JEV pathogenesis and transmission. In 3-week-old ICR mice, SA(14)-14-2 did not cause any symptoms or death after either intracerebral or peripheral inoculation with a maximum dose of up to 1.5×10(3) plaque-forming units (PFU) per mouse. The SA(14)-14-2 genome consisted of 10977 nucleotides, one nucleotide longer than all the previously reported genomes of SA(14)-14-2, SA(14) and two other SA(14)-derived attenuated viruses. This difference was due to an insertion of one G nucleotide at position 10701 in the 3 noncoding region. Also, we noted a significant number of nucleotide and/or amino acid substitutions throughout the genome of SA(14)-14-2, except for the prM protein-coding region, that differed from SA(14) and/or the other two attenuated viruses. Our results, together with others', provide a foundation not only for the study of JEV virulence but also for the development of new and improved vaccines for JEV.
Collapse
MESH Headings
- 3' Untranslated Regions
- Animals
- Cell Line
- Cricetinae
- Culicidae
- Disease Models, Animal
- Encephalitis Virus, Japanese/genetics
- Encephalitis Virus, Japanese/immunology
- Encephalitis Virus, Japanese/pathogenicity
- Encephalitis, Japanese/pathology
- Encephalitis, Japanese/prevention & control
- Encephalitis, Japanese/virology
- Female
- Genome, Viral
- Humans
- Japanese Encephalitis Vaccines/administration & dosage
- Japanese Encephalitis Vaccines/genetics
- Japanese Encephalitis Vaccines/immunology
- Mesocricetus
- Mice
- Mice, Inbred ICR
- Mutagenesis, Insertional
- Mutation, Missense
- RNA, Viral/genetics
- Time Factors
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Viral Plaque Assay
- Viral Proteins/genetics
- Virulence
Collapse
Affiliation(s)
- Byung-Hak Song
- Department of Microbiology, College of Medicine, Chungbuk National University, Cheongju 361-763, Republic of Korea
| | | | | | | | | |
Collapse
|
42
|
Lobigs M, Diamond MS. Feasibility of cross-protective vaccination against flaviviruses of the Japanese encephalitis serocomplex. Expert Rev Vaccines 2012; 11:177-87. [PMID: 22309667 DOI: 10.1586/erv.11.180] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Serological cross-reactivity providing cross-protective immunity between antigenically related viruses is a cornerstone of vaccination. It was the immunological basis for the first human vaccine against smallpox introduced more than 200 years ago, and continues to underpin modern vaccine development as has recently been shown for human papillomavirus vaccines, which confer cross-protection against other oncogenic papillomavirus types not present in the vaccine. Here, we review the feasibility of cross-protective vaccination against an antigenic group of clinically important viruses belonging to the Japanese encephalitis serocomplex in the Flaviviridae family. We will discuss evidence suggesting that 'new generation' flavivirus vaccines may provide effective cross-protective immunity against heterologous Japanese encephalitis serocomplex viruses, and appraise potential risks associated with cross-reactive vaccine immunity. The review will also focus on the structural and mechanistic basis for cross-protective immunity among this group of flaviviruses, which is predominantly mediated by antibodies against a single viral surface protein.
Collapse
Affiliation(s)
- Mario Lobigs
- Department of Emerging Pathogens & Vaccines, John Curtin School of Medical Research, The Australian National University, PO Box 334, Canberra, 2600, ACT, Australia.
| | | |
Collapse
|
43
|
Enzymatic analysis of recombinant Japanese encephalitis virus NS2B(H)-NS3pro protease with fluorogenic model peptide substrates. PLoS One 2012; 7:e36872. [PMID: 22615830 PMCID: PMC3352935 DOI: 10.1371/journal.pone.0036872] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 04/09/2012] [Indexed: 01/29/2023] Open
Abstract
Background Japanese encephalitis virus (JEV), a member of the Flaviviridae family, causes around 68,000 encephalitis cases annually, of which 20–30% are fatal, while 30–50% of the recovered cases develop severe neurological sequelae. Specific antivirals for JEV would be of great importance, particularly in those cases where the infection has become persistent. Being indispensable for flaviviral replication, the NS2B-NS3 protease is a promising target for design of anti-flaviviral inhibitors. Contrary to related flaviviral proteases, the JEV NS2B-NS3 protease is structurally and mechanistically much less characterized. Here we aimed at establishing a straightforward procedure for cloning, expression, purification and biochemical characterization of JEV NS2B(H)-NS3pro protease. Methodology/Principal Findings The full-length sequence of JEV NS2B-NS3 genotype III strain JaOArS 982 was obtained as a synthetic gene. The sequence of NS2B(H)-NS3pro was generated by splicing by overlap extension PCR (SOE-PCR) and cloned into the pTrcHisA vector. Hexahistidine-tagged NS2B(H)-NS3pro, expressed in E. coli as soluble protein, was purified to >95% purity by a single-step immobilized metal affinity chromatography. SDS-PAGE and immunoblotting of the purified enzyme demonstrated NS2B(H)-NS3pro precursor and its autocleavage products, NS3pro and NS2B(H), as 36, 21, and 10 kDa bands, respectively. Kinetic parameters, Km and kcat, for fluorogenic protease model substrates, Boc-GRR-amc, Boc-LRR-amc, Ac-nKRR-amc, Bz-nKRR-amc, Pyr-RTKR-amc and Abz-(R)4SAG-nY-amide, were obtained using inner filter effect correction. The highest catalytic efficiency kcat/Km was found for Pyr-RTKR-amc (kcat/Km: 1962.96±85.0 M−1 s−1) and the lowest for Boc-LRR-amc (kcat/Km: 3.74±0.3 M−1 s−1). JEV NS3pro is inhibited by aprotinin but to a lesser extent than DEN and WNV NS3pro. Conclusions/Significance A simplified procedure for the cloning, overexpression and purification of the NS2B(H)-NS3pro was established which is generally applicable to other flaviviral proteases. Kinetic parameters obtained for a number of model substrates and inhibitors, are useful for the characterization of substrate specificity and eventually for the design of high-throughput assays aimed at antiviral inhibitor discovery.
Collapse
|
44
|
Degrees of maturity: the complex structure and biology of flaviviruses. Curr Opin Virol 2012; 2:168-75. [PMID: 22445964 DOI: 10.1016/j.coviro.2012.02.011] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 02/20/2012] [Accepted: 02/22/2012] [Indexed: 11/21/2022]
Abstract
Flaviviruses are small enveloped virions that enter target cells in a pH-dependent fashion. Virus attachment, entry, and membrane fusion are orchestrated by the envelope (E) and pre-membrane (prM) proteins, the two structural proteins displayed on the surface of virions. Flaviviruses assemble as an immature non-infectious form onto which prM and E form trimeric spikes. During egress from infected cells, flaviviruses undergo dramatic structural changes characterized by the formation of a herringbone arrangement of E proteins that lie flat against the surface of the virion and cleavage of the prM protein by the cellular protease furin. The result is a relatively smooth, infectious mature virion. This dynamic process is now understood in structural detail at the atomic level. However, recent studies indicate that many of the virions released from cells share structural features of both immature and mature virus particles. These mosaic partially mature virions are infectious and interact uniquely with target cells and the host immune response. Here, we will discuss recent advances in our understanding of the biology and significance of partially mature flaviviruses.
Collapse
|
45
|
Tafuku S, Miyata T, Tadano M, Mitsumata R, Kawakami H, Harakuni T, Sewaki T, Arakawa T. Japanese encephalitis virus structural and nonstructural proteins expressed in Escherichia coli induce protective immunity in mice. Microbes Infect 2012; 14:169-76. [DOI: 10.1016/j.micinf.2011.09.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 09/22/2011] [Accepted: 09/25/2011] [Indexed: 02/09/2023]
|
46
|
Switzer WM, Zheng H, Simmons G, Zhou Y, Tang S, Shankar A, Kapusinszky B, Delwart EL, Heneine W. No evidence of murine leukemia virus-related viruses in live attenuated human vaccines. PLoS One 2011; 6:e29223. [PMID: 22216219 PMCID: PMC3245253 DOI: 10.1371/journal.pone.0029223] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 11/22/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The association of xenotropic murine leukemia virus (MLV)-related virus (XMRV) in prostate cancer and chronic fatigue syndrome reported in previous studies remains controversial as these results have been questioned by recent data. Nonetheless, concerns have been raised regarding contamination of human vaccines as a possible source of introduction of XMRV and MLV into human populations. To address this possibility, we tested eight live attenuated human vaccines using generic PCR for XMRV and MLV sequences. Viral metagenomics using deep sequencing was also done to identify the possibility of other adventitious agents. RESULTS All eight live attenuated vaccines, including Japanese encephalitis virus (JEV) (SA-14-14-2), varicella (Varivax), measles, mumps, and rubella (MMR-II), measles (Attenuvax), rubella (Meruvax-II), rotavirus (Rotateq and Rotarix), and yellow fever virus were negative for XMRV and highly related MLV sequences. However, residual hamster DNA, but not RNA, containing novel endogenous gammaretrovirus sequences was detected in the JEV vaccine using PCR. Metagenomics analysis did not detect any adventitious viral sequences of public health concern. Intracisternal A particle sequences closest to those present in Syrian hamsters and not mice were also detected in the JEV SA-14-14-2 vaccine. Combined, these results are consistent with the production of the JEV vaccine in Syrian hamster cells. CONCLUSIONS We found no evidence of XMRV and MLV in eight live attenuated human vaccines further supporting the safety of these vaccines. Our findings suggest that vaccines are an unlikely source of XMRV and MLV exposure in humans and are consistent with the mounting evidence on the absence of these viruses in humans.
Collapse
Affiliation(s)
- William M Switzer
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Emergence of genotype I of Japanese encephalitis virus as the dominant genotype in Asia. J Virol 2011; 85:9847-53. [PMID: 21697481 DOI: 10.1128/jvi.00825-11] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Japanese encephalitis virus (JEV), a mosquito-borne zoonotic pathogen, is one of the major causes of viral encephalitis worldwide. Previous phylogenetic studies based on the envelope protein indicated that there are four genotypes, and surveillance data suggest that genotype I is gradually replacing genotype III as the dominant strain. Here we report an evolutionary analysis based on 98 full-length genome sequences of JEV, including 67 new samples isolated from humans, pigs, mosquitoes, midges. and bats in affected areas. To investigate the relationships between the genotypes and the significance of genotype I in recent epidemics, we estimated evolutionary rates, ages of common ancestors, and population demographics. Our results indicate that the genotypes diverged in the order IV, III, II, and I and that the genetic diversity of genotype III has decreased rapidly while that of genotype I has increased gradually, consistent with its emergence as the dominant genotype.
Collapse
|
48
|
Pivotal role of antibody and subsidiary contribution of CD8+ T cells to recovery from infection in a murine model of Japanese encephalitis. J Virol 2011; 85:5446-55. [PMID: 21450826 DOI: 10.1128/jvi.02611-10] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The immunological correlates for recovery from primary Japanese encephalitis virus (JEV) infection in humans and experimental animals remain poorly defined. To investigate the relative importance of the adaptive immune responses, we have established a mouse model for Japanese encephalitis in which a low-dose virus inoculum was administered into the footpads of adult C57BL/6 mice. In this model, ~60% of the mice developed a fatal encephalitis and a virus burden in the central nervous system (CNS). Using mice lacking B cells (μMT(-/-) mice) and immune B cell transfer to wild-type mice, we show a critically important role for humoral immunity in preventing virus spread to the CNS. T cell help played an essential part in the maintenance of an effective antibody response necessary to combat the infection, since mice lacking major histocompatibility complex class II showed truncated IgM and blunted IgG responses and uniformly high lethality. JEV infection resulted in extensive CD8(+) T cell activation, judged by upregulation of surface markers CD69 and CD25 and cytokine production after stimulation with a JEV NS4B protein-derived H-2D(b)-binding peptide and trafficking of virus-immune CD8(+) T cells into the CNS. However, no significant effect of CD8(+) T cells on the survival phenotype was found, which was corroborated in knockout mice lacking key effector molecules (Fas receptor, perforin, or granzymes) of cytolytic pathways triggered by T lymphocytes. Accordingly, CD8(+) T cells are mostly dispensable for recovery from infection with JEV. This finding highlights the conflicting role that CD8(+) T cells play in the pathogenesis of JEV and closely related encephalitic flaviviruses such as West Nile virus.
Collapse
|
49
|
Mallilankaraman K, Shedlock DJ, Bao H, Kawalekar OU, Fagone P, Ramanathan AA, Ferraro B, Stabenow J, Vijayachari P, Sundaram SG, Muruganandam N, Sarangan G, Srikanth P, Khan AS, Lewis MG, Kim JJ, Sardesai NY, Muthumani K, Weiner DB. A DNA vaccine against chikungunya virus is protective in mice and induces neutralizing antibodies in mice and nonhuman primates. PLoS Negl Trop Dis 2011; 5:e928. [PMID: 21264351 PMCID: PMC3019110 DOI: 10.1371/journal.pntd.0000928] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 11/30/2010] [Indexed: 12/23/2022] Open
Abstract
Chikungunya virus (CHIKV) is an emerging mosquito-borne alphavirus indigenous to tropical Africa and Asia. Acute illness is characterized by fever, arthralgias, conjunctivitis, rash, and sometimes arthritis. Relatively little is known about the antigenic targets for immunity, and no licensed vaccines or therapeutics are currently available for the pathogen. While the Aedes aegypti mosquito is its primary vector, recent evidence suggests that other carriers can transmit CHIKV thus raising concerns about its spread outside of natural endemic areas to new countries including the U.S. and Europe. Considering the potential for pandemic spread, understanding the development of immunity is paramount to the development of effective counter measures against CHIKV. In this study, we isolated a new CHIKV virus from an acutely infected human patient and developed a defined viral challenge stock in mice that allowed us to study viral pathogenesis and develop a viral neutralization assay. We then constructed a synthetic DNA vaccine delivered by in vivo electroporation (EP) that expresses a component of the CHIKV envelope glycoprotein and used this model to evaluate its efficacy. Vaccination induced robust antigen-specific cellular and humoral immune responses, which individually were capable of providing protection against CHIKV challenge in mice. Furthermore, vaccine studies in rhesus macaques demonstrated induction of nAb responses, which mimicked those induced in convalescent human patient sera. These data suggest a protective role for nAb against CHIKV disease and support further study of envelope-based CHIKV DNA vaccines. Chikungunya fever epidemics are sustained by a cycle of human-mosquito-human transmission, with the epidemic cycle being similar to those of dengue and urban yellow fever. While the threat of a pandemic continues to engage the public's attention, the peculiar problems associated with the more immediate and very real seasonal epidemics are also worthy of consideration. Specifically, there are limited viral strains that have been characterized and available for laboratory study as well as limited knowledge of immune responses induced to the virus. In this study, we isolated CHIKV virus from an acutely infected human patient and used this new virus to develop a neutralization assay and a challenge stock, which is effective in a mouse model. Furthermore, we analyzed the ability of an envelope-based synthetic DNA-based vaccine to impact viral disease in the mouse model and to generate protective levels of immune responses in nonhuman primates. We observed that this novel vaccine approach generated protective levels of immune responses in both mouse and non-human primate models. We believe that these studies advance the field of Chikungunya vaccine research as well as the study of immune protection to CHIKV.
Collapse
Affiliation(s)
- Karthik Mallilankaraman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Devon J. Shedlock
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Huihui Bao
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Omkar U. Kawalekar
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Paolo Fagone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Aarthi A. Ramanathan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Bernadette Ferraro
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jennifer Stabenow
- Regional Biocontainment Lab, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Paluru Vijayachari
- Regional Medical Research Centers, Indian Council of Medical Research, Port Blair, Andaman & Nicobar Islands, India
| | - Senthil G. Sundaram
- Regional Medical Research Centers, Indian Council of Medical Research, Port Blair, Andaman & Nicobar Islands, India
| | - Nagarajan Muruganandam
- Regional Medical Research Centers, Indian Council of Medical Research, Port Blair, Andaman & Nicobar Islands, India
| | - Gopalsamy Sarangan
- Department of Microbiology, Sri Ramachandra Medical College & Research Institute, Chennai, India
| | - Padma Srikanth
- Department of Microbiology, Sri Ramachandra Medical College & Research Institute, Chennai, India
| | - Amir S. Khan
- Inovio Pharmaceuticals, Blue Bell, Pennsylvania, United States of America
| | - Mark G. Lewis
- Bioqual Inc, Rockville, Maryland, United States of America
| | - J. Joseph Kim
- Inovio Pharmaceuticals, Blue Bell, Pennsylvania, United States of America
| | | | - Karuppiah Muthumani
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - David B. Weiner
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
50
|
Li Y, Ye J, Yang X, Xu M, Chen L, Mei L, Zhu J, Liu X, Chen H, Cao S. Infection of mouse bone marrow-derived dendritic cells by live attenuated Japanese encephalitis virus induces cells maturation and triggers T cells activation. Vaccine 2011; 29:855-62. [DOI: 10.1016/j.vaccine.2010.09.108] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 09/02/2010] [Accepted: 09/26/2010] [Indexed: 10/18/2022]
|