1
|
Sann S, Kleinewietfeld M, Cantaert T. Balancing functions of regulatory T cells in mosquito-borne viral infections. Emerg Microbes Infect 2024; 13:2304061. [PMID: 38192073 PMCID: PMC10812859 DOI: 10.1080/22221751.2024.2304061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/07/2024] [Indexed: 01/10/2024]
Abstract
Mosquito-borne viral infections are on the rise worldwide and can lead to severe symptoms such as haemorrhage, encephalitis, arthritis or microcephaly. A protective immune response following mosquito-borne viral infections requires the generation of a controlled and balanced immune response leading to viral clearance without immunopathology. Here, regulatory T cells play a central role in restoring immune homeostasis. In current review, we aim to provide an overview and summary of the phenotypes of FOXP3+ Tregs in various mosquito-borne arboviral disease, their association with disease severity and their functional characteristics. Furthermore, we discuss the role of cytokines and Tregs in the immunopathogenesis of mosquito-borne infections. Lastly, we discuss possible novel lines of research which could provide additional insight into the role of Tregs in mosquito-borne viral infections in order to develop novel therapeutic approaches or vaccination strategies.
Collapse
Affiliation(s)
- Sotheary Sann
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Markus Kleinewietfeld
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), Hasselt University, Diepenbeek, Belgium
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| |
Collapse
|
2
|
Zhang W, Yu Q, Gao X, Chen H, Su J, Chen Y, Li Y, Zhang N, Fu Z, Cui M. Myeloid-Derived Suppressor Cells Induce Exhaustion-Like CD8 + T Cells during JEV Infection. Int J Biol Sci 2024; 20:5959-5978. [PMID: 39664572 PMCID: PMC11628328 DOI: 10.7150/ijbs.102372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/18/2024] [Indexed: 12/13/2024] Open
Abstract
Japanese encephalitis (JE), caused by Japanese encephalitis virus (JEV), is a mosquito-borne zoonotic disease and a leading cause of viral encephalitis worldwide. While JEV has the ability to traverse the blood-brain barrier (BBB), the precise mechanisms by which it inhibits the immune response prior to penetrating the BBB remain unclear, presenting obstacles in the development of efficacious therapeutic interventions. This study investigated the impact of JEV on CD8+ T cell responses, with a particular focus on the dysfunction of CD8+ T cells during JEV infection. Our results demonstrated that JEV infection significantly elevated the expression of PD-1 and TIM-3 on CD8+ T cells, which are markers of T cell exhaustion, leading to inhibited function and impaired differentiation, resulting in a poorer prognosis in mice. Compared with nondiseased mice, symptomatic mice presented a greater proportion of exhaustion-like CD8+ T cells. In vitro experiments further demonstrated that MDSCs induced an exhaustion-like state in CD8+ T cells, characterized by significant upregulation of PD-1 and TIM-3 expression. Notably, blocking TIM-3 or depleting MDSCs restored CD8+ T cell functionality by rescuing the expression of IFN-γ and TNF-α. Furthermore, the depletion of MDSCs not only alleviated T cell exhaustion-like phenotypes but also improved survival rates in JEV-infected mice. These findings suggest that JEV promotes immune evasion through MDSC-induced CD8+ T cell exhaustion-like states and identify TIM-3 as a promising therapeutic target for JE treatment.
Collapse
Affiliation(s)
- Weijia Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
- International Research Centre for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Qing Yu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
- International Research Centre for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Xiaochen Gao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
- International Research Centre for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Haowei Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
- International Research Centre for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Jie Su
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
- International Research Centre for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Yanru Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
- International Research Centre for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Yanling Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
- International Research Centre for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Nan Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
- International Research Centre for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Zhenfang Fu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
- International Research Centre for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| | - Min Cui
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
- International Research Centre for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, China
| |
Collapse
|
3
|
Pavlou A, Mulenge F, Gern OL, Busker LM, Greimel E, Waltl I, Kalinke U. Orchestration of antiviral responses within the infected central nervous system. Cell Mol Immunol 2024; 21:943-958. [PMID: 38997413 PMCID: PMC11364666 DOI: 10.1038/s41423-024-01181-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/05/2024] [Indexed: 07/14/2024] Open
Abstract
Many newly emerging and re-emerging viruses have neuroinvasive potential, underscoring viral encephalitis as a global research priority. Upon entry of the virus into the CNS, severe neurological life-threatening conditions may manifest that are associated with high morbidity and mortality. The currently available therapeutic arsenal against viral encephalitis is rather limited, emphasizing the need to better understand the conditions of local antiviral immunity within the infected CNS. In this review, we discuss new insights into the pathophysiology of viral encephalitis, with a focus on myeloid cells and CD8+ T cells, which critically contribute to protection against viral CNS infection. By illuminating the prerequisites of myeloid and T cell activation, discussing new discoveries regarding their transcriptional signatures, and dissecting the mechanisms of their recruitment to sites of viral replication within the CNS, we aim to further delineate the complexity of antiviral responses within the infected CNS. Moreover, we summarize the current knowledge in the field of virus infection and neurodegeneration and discuss the potential links of some neurotropic viruses with certain pathological hallmarks observed in neurodegeneration.
Collapse
Affiliation(s)
- Andreas Pavlou
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Felix Mulenge
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Olivia Luise Gern
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Lena Mareike Busker
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, 30559, Hannover, Germany
| | - Elisabeth Greimel
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Inken Waltl
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
4
|
Fan YC, Chen JM, Chen YY, Ke YD, Chang GJJ, Chiou SS. Epitope(s) involving amino acids of the fusion loop of Japanese encephalitis virus envelope protein is(are) important to elicit protective immunity. J Virol 2024; 98:e0177323. [PMID: 38530012 PMCID: PMC11019926 DOI: 10.1128/jvi.01773-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
Dengue vaccine candidates have been shown to improve vaccine safety and efficacy by altering the residues or accessibility of the fusion loop on the virus envelope protein domain II (DIIFL) in an ex vivo animal study. The current study aimed to comprehensively investigate the impact of DIIFL mutations on the antigenicity, immunogenicity, and protective efficacy of Japanese encephalitis virus (JEV) virus-like particles (VLPs) in mice. We found the DIIFL G106K/L107D (KD) and W101G/G106K/L107D (GKD) mutations altered the binding activity of JEV VLP to cross-reactive monoclonal antibodies but had no effect on their ability to elicit total IgG antibodies in mice. However, JEV VLPs with KD or GKD mutations induced significantly less neutralizing antibodies against JEV. Only 46% and 31% of the KD and GKD VLPs-immunized mice survived compared to 100% of the wild-type (WT) VLP-immunized mice after a lethal JEV challenge. In passive protection experiments, naïve mice that received sera from WT VLP-immunized mice exhibited a significantly higher survival rate of 46.7% compared to those receiving sera from KD VLP- and GKD VLP-immunized mice (6.7% and 0%, respectively). This study demonstrated that JEV DIIFL is crucial for eliciting potently neutralizing antibodies and protective immunity against JEV. IMPORTANCE Introduction of mutations into the fusion loop is one potential strategy for generating safe dengue and Zika vaccines by reducing the risk of severe dengue following subsequent infections, and for constructing live-attenuated vaccine candidates against newly emerging Japanese encephalitis virus (JEV) or Japanese encephalitis (JE) serocomplex virus. The monoclonal antibody studies indicated the fusion loop of JE serocomplex viruses primarily comprised non-neutralizing epitopes. However, the present study demonstrates that the JEV fusion loop plays a critical role in eliciting protective immunity in mice. Modifications to the fusion loop of JE serocomplex viruses might negatively affect vaccine efficacy compared to dengue and zika serocomplex viruses. Further studies are required to assess the impact of mutant fusion loop encoded by commonly used JEV vaccine strains on vaccine efficacy or safety after subsequent dengue virus infection.
Collapse
Affiliation(s)
- Yi-Chin Fan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Master of Public Health Degree Program, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Jo-Mei Chen
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Ying Chen
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yuan-Dun Ke
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Gwong-Jen J. Chang
- Arboviral Diseases Branch, Centers for Disease Control and Prevention, Fort, Fort Collins, Colorado, USA
| | - Shyan-Song Chiou
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
5
|
van Bree JW, Visser I, Duyvestyn JM, Aguilar-Bretones M, Marshall EM, van Hemert MJ, Pijlman GP, van Nierop GP, Kikkert M, Rockx BH, Miesen P, Fros JJ. Novel approaches for the rapid development of rationally designed arbovirus vaccines. One Health 2023; 16:100565. [PMID: 37363258 PMCID: PMC10288159 DOI: 10.1016/j.onehlt.2023.100565] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 06/28/2023] Open
Abstract
Vector-borne diseases, including those transmitted by mosquitoes, account for more than 17% of infectious diseases worldwide. This number is expected to rise with an increased spread of vector mosquitoes and viruses due to climate change and man-made alterations to ecosystems. Among the most common, medically relevant mosquito-borne infections are those caused by arthropod-borne viruses (arboviruses), especially members of the genera Flavivirus and Alphavirus. Arbovirus infections can cause severe disease in humans, livestock and wildlife. Severe consequences from infections include congenital malformations as well as arthritogenic, haemorrhagic or neuroinvasive disease. Inactivated or live-attenuated vaccines (LAVs) are available for a small number of arboviruses; however there are no licensed vaccines for the majority of these infections. Here we discuss recent developments in pan-arbovirus LAV approaches, from site-directed attenuation strategies targeting conserved determinants of virulence to universal strategies that utilize genome-wide re-coding of viral genomes. In addition to these approaches, we discuss novel strategies targeting mosquito saliva proteins that play an important role in virus transmission and pathogenesis in vertebrate hosts. For rapid pre-clinical evaluations of novel arbovirus vaccine candidates, representative in vitro and in vivo experimental systems are required to assess the desired specific immune responses. Here we discuss promising models to study attenuation of neuroinvasion, neurovirulence and virus transmission, as well as antibody induction and potential for cross-reactivity. Investigating broadly applicable vaccination strategies to target the direct interface of the vertebrate host, the mosquito vector and the viral pathogen is a prime example of a One Health strategy to tackle human and animal diseases.
Collapse
Affiliation(s)
- Joyce W.M. van Bree
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Imke Visser
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jo M. Duyvestyn
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, the Netherlands
| | | | - Eleanor M. Marshall
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Martijn J. van Hemert
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Gorben P. Pijlman
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | | | - Marjolein Kikkert
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Barry H.G. Rockx
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Pascal Miesen
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500, HB, Nijmegen, the Netherlands
| | - Jelke J. Fros
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| |
Collapse
|
6
|
Frank JC, Song BH, Lee YM. Mice as an Animal Model for Japanese Encephalitis Virus Research: Mouse Susceptibility, Infection Route, and Viral Pathogenesis. Pathogens 2023; 12:pathogens12050715. [PMID: 37242385 DOI: 10.3390/pathogens12050715] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Japanese encephalitis virus (JEV), a zoonotic flavivirus, is principally transmitted by hematophagous mosquitoes, continually between susceptible animals and incidentally from those animals to humans. For almost a century since its discovery, JEV was geographically confined to the Asia-Pacific region with recurrent sizable outbreaks involving wildlife, livestock, and people. However, over the past decade, it has been detected for the first time in Europe (Italy) and Africa (Angola) but has yet to cause any recognizable outbreaks in humans. JEV infection leads to a broad spectrum of clinical outcomes, ranging from asymptomatic conditions to self-limiting febrile illnesses to life-threatening neurological complications, particularly Japanese encephalitis (JE). No clinically proven antiviral drugs are available to treat the development and progression of JE. There are, however, several live and killed vaccines that have been commercialized to prevent the infection and transmission of JEV, yet this virus remains the main cause of acute encephalitis syndrome with high morbidity and mortality among children in the endemic regions. Therefore, significant research efforts have been directed toward understanding the neuropathogenesis of JE to facilitate the development of effective treatments for the disease. Thus far, multiple laboratory animal models have been established for the study of JEV infection. In this review, we focus on mice, the most extensively used animal model for JEV research, and summarize the major findings on mouse susceptibility, infection route, and viral pathogenesis reported in the past and present, and discuss some unanswered key questions for future studies.
Collapse
Affiliation(s)
- Jordan C Frank
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA
| | - Byung-Hak Song
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA
| | - Young-Min Lee
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA
| |
Collapse
|
7
|
Adam A, Lee C, Wang T. Rational Development of Live-Attenuated Zika Virus Vaccines. Pathogens 2023; 12:194. [PMID: 36839466 PMCID: PMC9963317 DOI: 10.3390/pathogens12020194] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Zika virus (ZIKV), a re-emerging mosquito-borne flavivirus, has caused outbreaks in Africa, Asia, the Pacific, and, more recently, in the Americas. ZIKV has been associated with the neurological autoimmune disorder Guillain-Barre syndrome in adults and congenital Zika syndrome in fetuses and infants, including microcephaly, spontaneous abortion, and intrauterine growth restriction. It is considered to be a major threat to global public health due to its unprecedented clinical impact on humans. Currently, there are no specific prophylactics or therapeutics available to prevent or treat ZIKV infection. The development of a safe and efficacious ZIKV vaccine remains a global health priority. Since the recent outbreak, multiple platforms have been used in the development of candidate ZIKV vaccines. The candidate vaccines have been shown to elicit strong T cell and neutralization antibody responses and protect against ZIKV infection in animal models. Some candidates have progressed successfully to clinical trials. Live-attenuated vaccines, which induce rapid and durable protective immunity, are one of the most important strategies for controlling flavivirus diseases. In this review, we discuss recent progress in the development of candidate live-attenuated ZIKV vaccines.
Collapse
Affiliation(s)
- Awadalkareem Adam
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Christy Lee
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
8
|
Mechanisms of Neuroinvasion and Neuropathogenesis by Pathologic Flaviviruses. Viruses 2023; 15:v15020261. [PMID: 36851477 PMCID: PMC9965671 DOI: 10.3390/v15020261] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/07/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Flaviviruses are present on every continent and cause significant morbidity and mortality. In many instances, severe cases of infection with flaviviruses involve the invasion of and damage to the central nervous system (CNS). Currently, there are several mechanisms by which it has been hypothesized flaviviruses reach the brain, including the disruption of the blood-brain barrier (BBB) which acts as a first line of defense by blocking the entry of many pathogens into the brain, passing through the BBB without disruption, as well as travelling into the CNS through axonal transport from peripheral nerves. After flaviviruses have entered the CNS, they cause different neurological symptoms, leading to years of neurological sequelae or even death. Similar to neuroinvasion, there are several identified mechanisms of neuropathology, including direct cell lysis, blockage of the cell cycle, indication of apoptosis, as well as immune induced pathologies. In this review, we aim to summarize the current knowledge in the field of mechanisms of both neuroinvasion and neuropathogenesis during infection with a variety of flaviviruses and examine the potential contributions and timing of each discussed pathway.
Collapse
|
9
|
Jiang S, Wu S, Zhao G, He Y, Bao L, Liu J, Qin C, Hou J, Ding Y, Cheng A, Jiang B, Wu J, Yan J, Humeau L, Patella A, Weiner DB, Broderick K, Wang B. Comparison of Wild Type DNA Sequence of Spike Protein from SARS-CoV-2 with Optimized Sequence on The Induction of Protective Responses Against SARS-Cov-2 Challenge in Mouse Model. Hum Vaccin Immunother 2022; 18:2016201. [PMID: 35061975 PMCID: PMC8986195 DOI: 10.1080/21645515.2021.2016201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Genetic optimization of Nucleic Acid immunogens is important for potentially improving their immune potency. A COVID-19 DNA vaccine is in phase III clinical trial which is based on a promising highly developable technology platform. Here, we show optimization in mice generating a pGX-9501 DNA vaccine encoding full-length spike protein, which results in induction of potent humoral and cellular immune responses, including neutralizing antibodies, that block hACE2-RBD binding of live CoV2 virus in vitro. Optimization resulted in improved induction of cellular immunity by pGX-9501 as demonstrated by increased IFN-γ expression in both CD8+ and CD4 + T cells and this was associated with more robust antiviral CTL responses compared to unoptimized constructs. Vaccination with pGX-9501 induced subsequent protection against virus challenge in a rigorous hACE2 transgenic mouse model. Overall, pGX-9501 is a promising optimized COVID-19 DNA vaccine candidate inducing humoral and cellular immunity contributing to the vaccine's protective effects.
Collapse
Affiliation(s)
- Sheng Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College (SHMC), Fudan University, Shanghai, China,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuting Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College (SHMC), Fudan University, Shanghai, China
| | - Gan Zhao
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - Yue He
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - Linlin Bao
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China
| | - Jiangning Liu
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China
| | - Chuan Qin
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China
| | - Jiawang Hou
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - Yuan Ding
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - Alex Cheng
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - Brian Jiang
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - John Wu
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - Jian Yan
- Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | | | - Ami Patella
- Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | | | | | - Bin Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College (SHMC), Fudan University, Shanghai, China,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China,CONTACT Bin Wang School of Basic Medical Sciences, Fudan University, 131 Dong’an Road, 409 Fuxing Building, Shanghai200032, China
| |
Collapse
|
10
|
Virulence and Cross-Protection Conferred by an Attenuated Genotype I-Based Chimeric Japanese Encephalitis Virus Strain Harboring the E Protein of Genotype V in Mice. Microbiol Spectr 2022; 10:e0199022. [PMID: 36301111 PMCID: PMC9769820 DOI: 10.1128/spectrum.01990-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Japanese encephalitis virus (JEV) genotype V (GV) emerged in China in 2009, then South Korea, and has since spread to other regions in Asia and beyond, raising concern about its pathogenicity and the cross-protection offered by JEV vaccines against different genotypes. In this study, we replaced the structural proteins (C-prM-E) of an attenuated genotype I (GI) SD12-F120 strain with those of a virulent GV XZ0934 strain to construct a recombinant chimeric GI-GV JEV (JEV-GI/V) strain to determine the role of the structural proteins in virulence and cross-protection. The recombinant chimeric virus was highly neurovirulent and neuroinvasive in mice. This demonstrated the determinant role of the structural proteins in the virulence of the GV strain. Intracerebral or intraperitoneal inoculation of mice with JEV-GI/V-E5 harboring a combination of substitutions (N47K, L107F, E138K, H123R, and I176R) in E protein, but not mutants containing single substitution of these residues, resulted in decreased or disappeared mortality, suggesting that these residues synergistically, but not individually, played a role in determining the neurovirulence and neuroinvasiveness of the GV strain. Immunization of mice with attenuated strain JEV-GI/V-E5 provided complete protection and induced high neutralizing antibody titers against parental strain JEV-GI/V, but partial cross-protection and low cross-neutralizing antibodies titers against the heterologous GI and GIII strains in mice, suggesting the reduced cross-protection of JEV vaccines among different genotypes. Overall, these findings suggested the essential role of the structural proteins in determination of the virulence of GV strain, and highlighted the need for a novel vaccine against this newly emerged strain. IMPORTANCE The GV JEV showed an increase in epidemic areas, which exhibited higher pathogenicity in mice than the prevalent GI and GIII strains. We replaced a recombinant chimeric GI-GV JEV (JEV-GI/V) strain to determine the role of the structural proteins in virulence and cross-protection. It was found that the essential role of the structural proteins is to determinethe virulence of the GV strain. It is also suggested that there is reduced cross-protection of JEV vaccines among different genotypes, which provides basic data for subsequent JEV prevention, control, and new vaccine development.
Collapse
|
11
|
Khare B, Kuhn RJ. The Japanese Encephalitis Antigenic Complex Viruses: From Structure to Immunity. Viruses 2022; 14:2213. [PMID: 36298768 PMCID: PMC9607441 DOI: 10.3390/v14102213] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/09/2022] Open
Abstract
In the last three decades, several flaviviruses of concern that belong to different antigenic groups have expanded geographically. This has resulted in the presence of often more than one virus from a single antigenic group in some areas, while in Europe, Africa and Australia, additionally, multiple viruses belonging to the Japanese encephalitis (JE) serogroup co-circulate. Morphological heterogeneity of flaviviruses dictates antibody recognition and affects virus neutralization, which influences infection control. The latter is further impacted by sequential infections involving diverse flaviviruses co-circulating within a region and their cross-reactivity. The ensuing complex molecular virus-host interplay leads to either cross-protection or disease enhancement; however, the molecular determinants and mechanisms driving these outcomes are unclear. In this review, we provide an overview of the epidemiology of four JE serocomplex viruses, parameters affecting flaviviral heterogeneity and antibody recognition, host immune responses and the current knowledge of the cross-reactivity involving JE serocomplex flaviviruses that leads to differential clinical outcomes, which may inform future preventative and therapeutic interventions.
Collapse
Affiliation(s)
- Baldeep Khare
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Richard J. Kuhn
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
12
|
Meng R, Yang B, Feng C, Huang J, Wang X, Zhang D. The difference in CD4+ T cell immunity between high- and low-virulence Tembusu viruses is mainly related to residues 151 and 304 in the envelope protein. Front Immunol 2022; 13:890263. [PMID: 36016955 PMCID: PMC9395619 DOI: 10.3389/fimmu.2022.890263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 07/20/2022] [Indexed: 11/22/2022] Open
Abstract
Tembusu virus (TMUV) can result in a severe disease affecting domestic ducks. The role of T cells in protection from TMUV infection and the molecular basis of T cell-mediated protection against TMUV remain largely uncharacterized. Here, we used the high-virulence TMUV strain Y and the low-virulence TMUV strain PS to investigate the protective role for TMUV-specific CD4+ and CD8+ T cells. When tested in a 5-day-old Pekin duck model, Y and PS induced comparable levels of neutralizing antibody, whereas Y elicited significantly stronger cellular immune response relative to PS. Using a duck adoptive transfer model, we showed that both CD4+ and CD8+ T cells provided significant protection from TMUV-related disease, with CD8+ T cell conferring more robust protection to recipient ducklings. For TMUV, CD4+ T cells mainly provided help for neutralizing antibody response, whereas CD8+ T cells mainly mediated viral clearance from infected tissues. The difference in T cell immunity between Y and PS was primarily attributed to CD4+ T cells; adoptive transfer of Y-specific CD4+ T cells resulted in significantly enhanced protective ability, neutralizing antibody response, and viral clearance from the brain relative to PS-specific CD4+ T cells. Further investigations with chimeric viruses, mutant viruses, and their parental viruses identified two mutations (T151A and R304M) in the envelope (E) protein that contributed significantly to TMUV-specific CD4+ T cell-mediated protective ability and neutralizing antibody response, with more beneficial effects being conferred by R304M. These data indicate T cell-mediated immunity is important for protection from disease, for viral clearance from tissues, and for the production of neutralizing antibodies, and that the difference in CD4+T cell immunity between high- and low-virulence TMUV strains is primarily related to residues 151 and 304 in the E protein.
Collapse
|
13
|
Zhou Y, Bian P, Du H, Wang T, Li M, Hu H, Ye C, Zheng X, Zhang Y, Lei Y, Jia Z, Lian J. The Comparison of Inflammatory Cytokines (IL-6 and IL-18) and Immune Cells in Japanese Encephalitis Patients With Different Progression. Front Cell Infect Microbiol 2022; 12:826603. [PMID: 35463639 PMCID: PMC9022626 DOI: 10.3389/fcimb.2022.826603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/08/2022] [Indexed: 12/04/2022] Open
Abstract
Background Japanese encephalitis virus (JEV) is the main cause of viral encephalitis in Asia. Nowadays, no effective and specific therapy for JE patients is available except supportive treatment. The fatality rate of JE patients is still about 30%, and more than half of survivors suffered from various neuropsychiatric sequelae. Thus, more attention should be paid to JE. Methods In this study, a retrospective cohort of JE patients was collected and the general features of JE patients admitted into the Department of Infectious Diseases were analyzed. Meanwhile, the dynamic change of plasma cytokines and immune cells in JE patients with divergent prognosis was detected and analyzed. Results We found a mounted proportion of adult/old patients in JE cases. The level of IL-6 and IL-18 increased in JE patients especially in fatal individuals. There was a continuous decreased percentage of CD4+ T and B cells in severe JE patients with fatal outcome compared with the surviving JE patients. Conclusions The consistent high level of IL-6 and IL-18 in the plasma and low proportion of CD4+ T and B cells in the PBMCs might be the indicators of poor prognosis.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Peiyu Bian
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Hong Du
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Tao Wang
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Mengyuan Li
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Haifeng Hu
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Chuantao Ye
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Xuyang Zheng
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Ying Zhang
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Yinfeng Lei
- Department of Microbiology, School of Preclinical Medicine, Air Force Medical University, Xi’an, China
| | - Zhansheng Jia
- The Center of Infectious Diseases and Liver, Xi’an International Medical Center Hospital, Xi’an, China
- *Correspondence: Zhansheng Jia, ; Jianqi Lian,
| | - Jianqi Lian
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi’an, China
- *Correspondence: Zhansheng Jia, ; Jianqi Lian,
| |
Collapse
|
14
|
Jiang S, Wu S, Zhao G, He Y, Guo X, Zhang Z, Hou J, Ding Y, Cheng A, Wang B. Identification of a promiscuous conserved CTL epitope within the SARS-CoV-2 spike protein. Emerg Microbes Infect 2022; 11:730-740. [PMID: 35171086 PMCID: PMC8890520 DOI: 10.1080/22221751.2022.2043727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The COVID-19 disease caused by infection with SARS-CoV-2 and its variants is devastating to the global public health and economy. To date, over a hundred COVID-19 vaccines are known to be under development, and the few that have been approved to fight the disease are using the spike protein as the primary target antigen. Although virus-neutralizing epitopes are mainly located within the RBD of the spike protein, the presence of T cell epitopes, particularly the CTL epitopes that are likely to be needed for killing infected cells, has received comparatively little attention. This study predicted several potential T cell epitopes with web-based analytic tools and narrowed them down from several potential MHC-I and MHC-II epitopes by ELIspot and cytolytic assays to a conserved MHC-I epitope. The epitope is highly conserved in current viral variants and compatible with a presentation by most HLA alleles worldwide. In conclusion, we identified a CTL epitope suitable for evaluating the CD8+ T cell-mediated cellular response and potentially for addition into future COVID-19 vaccine candidates to maximize CTL responses against SARS-CoV-2.
Collapse
Affiliation(s)
- Sheng Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College(SHMC), Fudan University.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuting Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College(SHMC), Fudan University
| | - Gan Zhao
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| | - Yue He
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| | | | - Zhiyu Zhang
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| | - Jiawang Hou
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| | - Yuan Ding
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| | - Alex Cheng
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| | - Bin Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College(SHMC), Fudan University.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| |
Collapse
|
15
|
Sharma KB, Vrati S, Kalia M. Pathobiology of Japanese encephalitis virus infection. Mol Aspects Med 2021; 81:100994. [PMID: 34274157 DOI: 10.1016/j.mam.2021.100994] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 12/25/2022]
Abstract
Japanese encephalitis virus (JEV) is a flavivirus, spread by the bite of carrier Culex mosquitoes. The subsequent disease caused is Japanese encephalitis (JE), which is the leading global cause of virus-induced encephalitis. The disease is predominant in the entire Asia-Pacific region with the potential of global spread. JEV is highly neuroinvasive with symptoms ranging from mild fever to severe encephalitis and death. One-third of JE infections are fatal, and half of the survivors develop permanent neurological sequelae. Disease prognosis is determined by a series of complex and intertwined signaling events dictated both by the virus and the host. All flaviviruses, including JEV replicate in close association with ER derived membranes by channelizing the protein and lipid components of the ER. This leads to activation of acute stress responses in the infected cell-oxidative stress, ER stress, and autophagy. The host innate immune and inflammatory responses also enter the fray, the components of which are inextricably linked to the cellular stress responses. These are especially crucial in the periphery for dendritic cell maturation and establishment of adaptive immunity. The pathogenesis of JEV is a combination of direct virus induced neuronal cell death and an uncontrolled neuroinflammatory response. Here we provide a comprehensive review of the JEV life cycle and how the cellular stress responses dictate the pathobiology and resulting immune response. We also deliberate on how modulation of these stress pathways could be a potential strategy to develop therapeutic interventions, and define the persisting challenges.
Collapse
Affiliation(s)
- Kiran Bala Sharma
- Virology Research Group, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India
| | - Sudhanshu Vrati
- Virology Research Group, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India.
| | - Manjula Kalia
- Virology Research Group, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India.
| |
Collapse
|
16
|
Chauhan S, Rathore DK, Sachan S, Lacroix-Desmazes S, Gupta N, Awasthi A, Vrati S, Kalia M. Japanese Encephalitis Virus Infected Human Monocyte-Derived Dendritic Cells Activate a Transcriptional Network Leading to an Antiviral Inflammatory Response. Front Immunol 2021; 12:638694. [PMID: 34220803 PMCID: PMC8247639 DOI: 10.3389/fimmu.2021.638694] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 05/04/2021] [Indexed: 12/29/2022] Open
Abstract
A comprehensive understanding of the human immune response to virus infection is imperative for developing effective therapies, antivirals, and vaccines. Dendritic cells (DCs) are among the first cells to encounter the virus and are also key antigen-presenting cells that link the innate and adaptive immune system. In this study, we focus on the human immune response to the mosquito-borne Japanese encephalitis virus (JEV), which is the leading cause of virus-induced encephalitis in south-east Asia and has the potential to become a global pathogen. We describe the gene regulatory circuit of JEV infection in human monocyte-derived DCs (moDCs) along with its functional validation. We observe that JEV can productively infect human moDCs leading to robust transcriptional activation of the interferon and NF-κB-mediated antiviral and inflammatory pathways. This is accompanied with DC maturation and release of pro-inflammatory cytokines and chemokines TNFα, IL-6, IL-8, IL-12, MCP-1. and RANTES. JEV-infected moDCs activated T-regulatory cells (Tregs) in allogenic mixed lymphocyte reactions (MLR) as seen by upregulated FOXP3 mRNA expression, suggestive of a host response to reduce virus-induced immunopathology. The virus also downregulated transcripts involved in Peroxisome Proliferator Activated Receptor (PPAR) signalling and fatty acid metabolism pathways suggesting that changes in cellular metabolism play a crucial role in driving the DC maturation and antiviral responses. Collectively, our data describe and corroborate the human DC transcriptional network that is engaged upon JEV sensing.
Collapse
Affiliation(s)
| | | | - Shilpa Sachan
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, India
| | - Sebastien Lacroix-Desmazes
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| | - Nimesh Gupta
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, India
| | - Amit Awasthi
- Translational Health Science & Technology Institute, Faridabad, India
| | - Sudhanshu Vrati
- Translational Health Science & Technology Institute, Faridabad, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Manjula Kalia
- Translational Health Science & Technology Institute, Faridabad, India
- Regional Centre for Biotechnology, Faridabad, India
| |
Collapse
|
17
|
Bhattacharjee A, Chaudhuri R, Dash JJ, Saha M, Choudhury L, Roy S. Pre-treatment with Scopolamine Naturally Suppresses Japanese Encephalitis Viral Load in Embryonated Chick Through Regulation of Multiple Signaling Pathways. Appl Biochem Biotechnol 2021; 193:1654-1674. [PMID: 33620666 DOI: 10.1007/s12010-021-03526-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/07/2021] [Indexed: 11/30/2022]
Abstract
Suitable recognition of invasive microorganisms is a crucial factor for evoking a strong immune response that can combat the pathogen. Toll-like receptors (TLRs) play a pivotal role in the induction of this innate immune response through stimulation of interferons (IFNs) that control viral replication in the host via distinct signaling pathways. Though the antiviral property of Atropa belladonna has been established, yet the role of one of its active components scopolamine in modulating various factors of the innate immune branch has not yet been investigated until date. Thus, the present study was conducted to assess the antiviral effects of scopolamine and its immunomodulatory role against Japanese encephalitis virus (JEV) infections in embryonated chick. Pre-treatment with scopolamine hydrobromide showed a significant decrease in the viral loads of chorioallantoic membrane (CAM) and brain tissues. Molecular docking analysis revealed that scopolamine hydrobromide binds to the active site of non-structural protein 5 (NS5) that has enzymatic activities required for replication of JEV, making it a highly promising chemical compound against the virus. The binding contributions of different amino acid residues at or near the active site suggest a potential binding of this compound. Pre-treatment with the scopolamine hydrobromide showed significant upregulation of different TLRs like TLR3, TLR7, and TLR8, interleukins like IL-4, and IL-10, as well as IFNs and their regulatory factors. However, virus-infected tissues (direct infection group) exhibited higher TLR4 expression as compared to scopolamine hydrobromide pre-treated, virus-infected tissues (medicine pre-treated group). These results indicate that scopolamine hydrobromide contributes much to launch antiviral effects by remoulding the TLR and IFN signaling pathways that are involved in sensing and initiating the much-needed anti-JEV responses.
Collapse
Affiliation(s)
- Arghyadeep Bhattacharjee
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India
| | - Rajarshi Chaudhuri
- Department of Biotechnology, Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Jiban Jyoti Dash
- Department of Botany, Berhampur University, Berhampur, Odisha, India
| | - Manish Saha
- Department of Cardiology, R.G Kar Medical College & Hospital, Kolkata, West Bengal, India
| | | | - Souvik Roy
- Post-Graduate Department of Biotechnology, St. Xavier's College (Autonomous), Kolkata, West Bengal, India.
| |
Collapse
|
18
|
Adam A, Fontes-Garfias CR, Sarathy VV, Liu Y, Luo H, Davis E, Li W, Muruato AE, Wang B, Ahatov R, Mahmoud Y, Shan C, Osman SR, Widen SG, Barrett ADT, Shi PY, Wang T. A genetically stable Zika virus vaccine candidate protects mice against virus infection and vertical transmission. NPJ Vaccines 2021; 6:27. [PMID: 33597526 PMCID: PMC7889622 DOI: 10.1038/s41541-021-00288-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 01/15/2021] [Indexed: 12/20/2022] Open
Abstract
Although live attenuated vaccines (LAVs) have been effective in the control of flavivirus infections, to date they have been excluded from Zika virus (ZIKV) vaccine trials due to safety concerns. We have previously reported two ZIKV mutants, each of which has a single substitution in either envelope (E) glycosylation or nonstructural (NS) 4B P36 and displays a modest reduction in mouse neurovirulence and neuroinvasiveness, respectively. Here, we generated a ZIKV mutant, ZE4B-36, which combines mutations in both E glycosylation and NS4B P36. The ZE4B-36 mutant is stable and attenuated in viral replication. Next-generation sequence analysis showed that the attenuating mutations in the E and NS4B proteins are retained during serial cell culture passages. The mutant exhibits a significant reduction in neuroinvasiveness and neurovirulence and low infectivity in mosquitoes. It induces robust ZIKV-specific memory B cell, antibody, and T cell-mediated immune responses in type I interferon receptor (IFNR) deficient mice. ZIKV-specific T cell immunity remains strong months post-vaccination in wild-type C57BL/6 (B6) mice. Vaccination with ZE4B-36 protects mice from ZIKV-induced diseases and vertical transmission. Our results suggest that combination mutations in E glycosylation and NS4B P36 contribute to a candidate LAV with significantly increased safety but retain strong immunogenicity for prevention and control of ZIKV infection.
Collapse
Affiliation(s)
- Awadalkareem Adam
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Camila R Fontes-Garfias
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Vanessa V Sarathy
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Yang Liu
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Huanle Luo
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Emily Davis
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Wenqian Li
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Antonio E Muruato
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Binbin Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Renat Ahatov
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yoseph Mahmoud
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Chao Shan
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Samantha R Osman
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Steven G Widen
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
- Molecular Genomics Core Facility, University of Texas Medical Branch, Galveston, TX, USA
| | - Alan D T Barrett
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Pei-Yong Shi
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA.
- Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA.
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA.
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
19
|
Kalia A, Agrawal M, Gupta N. CD8 + T cells are crucial for humoral immunity establishment by SA14-14-2 live attenuated Japanese encephalitis vaccine in mice. Eur J Immunol 2020; 51:368-379. [PMID: 32749679 DOI: 10.1002/eji.202048745] [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: 05/12/2020] [Revised: 07/06/2020] [Indexed: 11/09/2022]
Abstract
The live attenuated SA14-14-2 Japanese encephalitis (JE) vaccine is a historical vaccine that protects against JE. Despite its extensive use, the mechanism of protective immunity conferred by the SA14-14-2 vaccine is not well established. Here, we used mouse models to understand the mechanism of the development of humoral immunity against the vaccine. The vaccine induces robust GC responses within a week postimmunization. In lethal virus challenge, we show that CD4+ T cells alone, but not CD8+ T cells, are sufficient to confer vaccine-mediated protection. However, the CD4-mediated protection was potentiated in the presence of vaccine-primed CD8+ T cells. Employing CD8-deficient mice, we show that both the protective traits of CD4+ T cells and the quality of antibody response to the vaccine are impaired in absence of CD8+ T cells. We further demonstrate that the poor protective immune response induced by the vaccine in absence of CD8+ T cells is mainly due to the impaired differentiation and function of follicular Th cells, leading to suboptimal GC reaction. Our study highlights an unprecedented role of CD8+ T cells in the establishment of humoral responses to the vaccine. By elucidating underlying cellular determinants of vaccine-induced protective immunity, our work has implications for rational design of vaccines against JE virus and related flaviviruses.
Collapse
Affiliation(s)
- Anurag Kalia
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, India
| | - Mona Agrawal
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, India
| | - Nimesh Gupta
- Vaccine Immunology Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
20
|
Fu TL, Ong KC, Tan SH, Wong KT. Japanese Encephalitis Virus Infects the Thalamus Early Followed by Sensory-Associated Cortex and Other Parts of the Central and Peripheral Nervous Systems. J Neuropathol Exp Neurol 2020; 78:1160-1170. [PMID: 31675093 DOI: 10.1093/jnen/nlz103] [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] [Indexed: 12/13/2022] Open
Abstract
Japanese encephalitis (JE) is a known CNS viral infection that often involves the thalamus early. To investigate the possible role of sensory peripheral nervous system (PNS) in early neuroinvasion, we developed a left hindlimb footpad-inoculation mouse model to recapitulate human infection by a mosquito bite. A 1-5 days postinfection (dpi) study, demonstrated focal viral antigens/RNA in contralateral thalamic neurons at 3 dpi in 50% of the animals. From 4 to 5 dpi, gradual increase in viral antigens/RNA was observed in bilateral thalami, somatosensory, and piriform cortices, and then the entire CNS. Infection of neuronal bodies and adjacent nerves in dorsal root ganglia (DRGs), trigeminal ganglia, and autonomic ganglia (intestine, etc.) was also observed from 5 dpi. Infection of explant organotypic whole brain slice cultures demonstrated no viral predilection for the thalamus, while DRG and intestinal ganglia organotypic cultures confirmed sensory and autonomic ganglia susceptibility to infection, respectively. Early thalamus and sensory-associated cortex involvement suggest an important role for sensory pathways in neuroinvasion. Our results suggest that JE virus neuronotropism is much more extensive than previously known, and that the sensory PNS and autonomic system are susceptible to infection.
Collapse
Affiliation(s)
- Tzeh Long Fu
- Department of Pathology; and Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Kien Chai Ong
- Department of Pathology; and Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Soon Hao Tan
- Department of Pathology; and Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Kum Thong Wong
- Department of Pathology; and Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
21
|
Sethi M, Das T, Tomar N, John JK, Dubal ZB, Rajak KK, Singh R, Saikumar G. Japanese encephalitis virus-induced neuropathology in mouse model infected through the conjunctival route. Indian J Med Res 2020; 150:498-503. [PMID: 31939394 PMCID: PMC6977363 DOI: 10.4103/ijmr.ijmr_2078_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background & objectives: Mouse is a preferred animal model for studying pathogenesis of Japanese encephalitis virus (JEV) infections, and different routes of inoculation have been tried. Some neurotropic viruses can reach the brain following infection through ocular route. This study was undertaken to establish JEV-induced clinical disease in mouse model through conjunctival route and document the neuropathological effects. Methods: Ten two-week old Swiss albino mice were inoculated with 5 μl Vero cell cultured virus containing 104.7 TCID50 JEV through conjunctival route. Clinical signs of mice were observed twice daily. After necropsy examination, different organs including eyes and olfactory bulbs were collected for histopathological examination, quantification of viral copy number and antigen by real-time TaqMan assay and immunohistochemistry, respectively. Results: Infected mice showed characteristic clinical signs of JE by 4 days post-infection (dpi). Histopathological lesions in brain included perivascular cuffing by mononuclear cells, focal gliosis, necrosis of neurons and neuronophagia and astrocytosis in the cerebrum, cerebellum and the brainstem. JEV viral load was highest in the brain followed by intestine, heart, liver, spleen, lung and kidney. JEV antigen was detected in the bipolar and ganglion cells of the retina and in the mitral cells and periglomerular cells of olfactory bulb and other parts of the brain. Interpretation & conclusions: JEV infection in mice through conjunctival route produced characteristic clinical signs of the disease and neuropathological lesions. Demonstration of JEV antigen in association with neuropathological lesions in the central nervous system and neuronal cells of the eye showed that conjunctival route could be an effective alternate route for virus invasion into the brain. These findings have biosafety implications for researchers, veterinary practitioners and pig farmers.
Collapse
Affiliation(s)
- Menaka Sethi
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Tareni Das
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Neelam Tomar
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Jeny K John
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Zunjar B Dubal
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Kaushal K Rajak
- Division of Biological Products, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Rajendra Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - G Saikumar
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| |
Collapse
|
22
|
Subramaniam KS, Lant S, Goodwin L, Grifoni A, Weiskopf D, Turtle L. Two Is Better Than One: Evidence for T-Cell Cross-Protection Between Dengue and Zika and Implications on Vaccine Design. Front Immunol 2020; 11:517. [PMID: 32269575 PMCID: PMC7109261 DOI: 10.3389/fimmu.2020.00517] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
Abstract
Dengue virus (DENV, family Flaviviridae, genus Flavivirus) exists as four distinct serotypes. Generally, immunity after infection with one serotype is protective and lifelong, though exceptions have been described. However, secondary infection with a different serotype can result in more severe disease for a minority of patients. Host responses to the first DENV infection involve the development of both cross-reactive antibody and T cell responses, which, depending upon their precise balance, may mediate protection or enhance disease upon secondary infection with a different serotype. Abundant evidence now exists that responses elicited by DENV infection can cross-react with other members of the genus Flavivirus, particularly Zika virus (ZIKV). Cohort studies have shown that prior DENV immunity is associated with protection against Zika. Cross-reactive antibody responses may enhance infection with flaviviruses, which likely accounts for the cases of severe disease seen during secondary DENV infections. Data for T cell responses are contradictory, and even though cross-reactive T cell responses exist, their clinical significance is uncertain. Recent mouse experiments, however, show that cross-reactive T cells are capable of mediating protection against ZIKV. In this review, we summarize and discuss the evidence that T cell responses may, at least in part, explain the cross-protection seen against ZIKV from DENV infection, and that T cell antigens should therefore be included in putative Zika vaccines.
Collapse
Affiliation(s)
- Krishanthi S. Subramaniam
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Suzannah Lant
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Lynsey Goodwin
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Alba Grifoni
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Daniela Weiskopf
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Lance Turtle
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
- Tropical and Infectious Disease Unit, Liverpool University Hospitals, Liverpool, United Kingdom
| |
Collapse
|
23
|
Chang YH, Chiao DJ, Hsu YL, Lin CC, Wu HL, Shu PY, Chang SF, Chang JH, Kuo SC. Mosquito Cell-Derived Japanese Encephalitis Virus-Like Particles Induce Specific Humoral and Cellular Immune Responses in Mice. Viruses 2020; 12:v12030336. [PMID: 32204533 PMCID: PMC7150764 DOI: 10.3390/v12030336] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/12/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
The Japanese encephalitis virus (JEV) is the major cause of an acute encephalitis syndrome in many Asian countries, despite the fact that an effective vaccine has been developed. Virus-like particles (VLPs) are self-assembled multi-subunit protein structures which possess specific epitope antigenicities related to corresponding native viruses. These properties mean that VLPs are considered safe antigens that can be used in clinical applications. In this study, we developed a novel baculovirus/mosquito (BacMos) expression system which potentially enables the scalable production of JEV genotype III (GIII) VLPs (which are secreted from mosquito cells). The mosquito-cell-derived JEV VLPs comprised 30-nm spherical particles as well as precursor membrane protein (prM) and envelope (E) proteins with densities that ranged from 30% to 55% across a sucrose gradient. We used IgM antibody-capture enzyme-linked immunosorbent assays to assess the resemblance between VLPs and authentic virions and thereby characterized the epitope specific antigenicity of VLPs. VLP immunization was found to elicit a specific immune response toward a balanced IgG2a/IgG1 ratio. This response effectively neutralized both JEV GI and GIII and elicited a mixed Th1/Th2 response in mice. This study supports the development of mosquito cell-derived JEV VLPs to serve as candidate vaccines against JEV.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Cell Line
- Culicidae/virology
- Cytokines/metabolism
- Disease Models, Animal
- Encephalitis Virus, Japanese/immunology
- Encephalitis Virus, Japanese/ultrastructure
- Encephalitis, Japanese/immunology
- Encephalitis, Japanese/virology
- Enzyme-Linked Immunosorbent Assay
- Epitopes/immunology
- Fluorescent Antibody Technique
- Immunity, Cellular
- Immunity, Humoral
- Mice
- Neutralization Tests
- Vaccines, Virus-Like Particle/immunology
- Virion
Collapse
Affiliation(s)
- Yu-Hsiu Chang
- Institute of Preventive Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-H.C.); (D.-J.C.); (Y.-L.H.); (C.-C.L.); (H.-L.W.); (J.-H.C.)
| | - Der-Jiang Chiao
- Institute of Preventive Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-H.C.); (D.-J.C.); (Y.-L.H.); (C.-C.L.); (H.-L.W.); (J.-H.C.)
| | - Yu-Lin Hsu
- Institute of Preventive Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-H.C.); (D.-J.C.); (Y.-L.H.); (C.-C.L.); (H.-L.W.); (J.-H.C.)
| | - Chang-Chi Lin
- Institute of Preventive Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-H.C.); (D.-J.C.); (Y.-L.H.); (C.-C.L.); (H.-L.W.); (J.-H.C.)
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 11490, Taiwan
| | - Hsueh-Ling Wu
- Institute of Preventive Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-H.C.); (D.-J.C.); (Y.-L.H.); (C.-C.L.); (H.-L.W.); (J.-H.C.)
| | - Pei-Yun Shu
- Center for Diagnostics and Vaccine Development, Centers for Disease Control, Ministry of Health and Welfare, Taipei 11561, Taiwan; (P.-Y.S.); (S.-F.C.)
| | - Shu-Fen Chang
- Center for Diagnostics and Vaccine Development, Centers for Disease Control, Ministry of Health and Welfare, Taipei 11561, Taiwan; (P.-Y.S.); (S.-F.C.)
| | - Jui-Huan Chang
- Institute of Preventive Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-H.C.); (D.-J.C.); (Y.-L.H.); (C.-C.L.); (H.-L.W.); (J.-H.C.)
| | - Szu-Cheng Kuo
- Institute of Preventive Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-H.C.); (D.-J.C.); (Y.-L.H.); (C.-C.L.); (H.-L.W.); (J.-H.C.)
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 11490, Taiwan
- Correspondence: ; Tel.: +886-2-8177-7038 (ext. 19946)
| |
Collapse
|
24
|
Saxena SK, Kumar S, Haikerwal A. Animal Flaviviruses. EMERGING AND TRANSBOUNDARY ANIMAL VIRUSES 2020. [DOI: 10.1007/978-981-15-0402-0_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
25
|
Li G, Adam A, Luo H, Shan C, Cao Z, Fontes-Garfias CR, Sarathy VV, Teleki C, Winkelmann ER, Liang Y, Sun J, Bourne N, Barrett ADT, Shi PY, Wang T. An attenuated Zika virus NS4B protein mutant is a potent inducer of antiviral immune responses. NPJ Vaccines 2019; 4:48. [PMID: 31815005 PMCID: PMC6883050 DOI: 10.1038/s41541-019-0143-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022] Open
Abstract
Live attenuated vaccines (LAVs) are one of the most important strategies to control flavivirus diseases. The flavivirus nonstructural (NS) 4B proteins are a critical component of both the virus replication complex and evasion of host innate immunity. Here we have used site-directed mutagenesis of residues in the highly conserved N-terminal and central hydrophobic regions of Zika virus (ZIKV) NS4B protein to identify candidate attenuating mutations. Three single-site mutants were generated, of which the NS4B-C100S mutant was more attenuated than the other two mutants (NS4B-C100A and NS4B-P36A) in two immunocompromised mouse models of fatal ZIKV disease. The ZIKV NS4B-C100S mutant triggered stronger type 1 interferons and interleukin-6 production, and higher ZIKV-specific CD4+ and CD8+ T-cell responses, but induced similar titers of neutralization antibodies compared with the parent wild-type ZIKV strain and a previously reported candidate ZIKV LAV with a 10-nucleotide deletion in 3'-UTR (ZIKV-3'UTR-Δ10). Vaccination with ZIKV NS4B-C100S protected mice from subsequent WT ZIKV challenge. Furthermore, either passive immunization with ZIKV NS4B-C100S immune sera or active immunization with ZIKV NS4B-C100S followed by the depletion of T cells affords full protection from lethal WT ZIKV challenge. In summary, our results suggest that the ZIKV NS4B-C100S mutant may serve as a candidate ZIKV LAV due to its attenuated phenotype and high immunogenicity.
Collapse
Affiliation(s)
- Guangyu Li
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Awadalkareem Adam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Huanle Luo
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Chao Shan
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Zengguo Cao
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Camila R. Fontes-Garfias
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Vanessa V. Sarathy
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Cody Teleki
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Evandro R. Winkelmann
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Nigel Bourne
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555 USA
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Alan D. T. Barrett
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Tian Wang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555 USA
| |
Collapse
|
26
|
Abstract
Japanese encephalitis (JE) is a clinical manifestation of the brain inflammation caused by JE virus (JEV). This virus imparts permanent neurological damage, thus imposing a heavy burden on public health and society. Neuro-inflammation is the hallmark of JEV infection. The prolonged pro-inflammatory response is due primarily to microglial activation, which eventually leads to severe encephalitis. A continual effort is going on in the scientific community toward an understanding of cellular and molecular factors that are involved in JEV neuro-invasion and inflammatory processes. This review not only gives a comprehensive update on the recent advances on understanding virus structure and mechanisms of pathogenesis but also briefly discusses crucial unresolved issues. We also highlight challenging areas of research that might open new avenues for controlling virus-induced neuro-inflammation.
Collapse
Affiliation(s)
- Arup Banerjee
- Laboratory of Virology, Regional Centre for Biotechnology, Faridabad, Haryana, India.,Translational Health Science & Technology Institute, Faridabad, Haryana, India
| | - Aarti Tripathi
- Translational Health Science & Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
27
|
Abstract
Japanese encephalitis is a mosquito-borne disease that occurs in Asia and is caused by Japanese encephalitis virus (JEV), a member of the genus Flavivirus. Although many flaviviruses can cause encephalitis, JEV causes particularly severe neurological manifestations. The virus causes loss of more disability-adjusted life years than any other arthropod-borne virus owing to the frequent neurological sequelae of the condition. Despite substantial advances in our understanding of Japanese encephalitis from in vitro studies and animal models, studies of pathogenesis and treatment in humans are lagging behind. Few mechanistic studies have been conducted in humans, and only four clinical trials of therapies for Japanese encephalitis have taken place in the past 10 years despite an estimated incidence of 69,000 cases per year. Previous trials for Japanese encephalitis might have been too small to detect important benefits of potential treatments. Many potential treatment targets exist for Japanese encephalitis, and pathogenesis and virological studies have uncovered mechanisms by which these drugs could work. In this Review, we summarize the epidemiology, clinical features, prevention and treatment of Japanese encephalitis and focus on potential new therapeutic strategies, based on repurposing existing compounds that are already suitable for human use and could be trialled without delay. We use our newly improved understanding of Japanese encephalitis pathogenesis to posit potential treatments and outline some of the many challenges that remain in tackling the disease in humans.
Collapse
|
28
|
CD4 T Cells, CD8 T Cells, and Monocytes Coordinate To Prevent Rift Valley Fever Virus Encephalitis. J Virol 2018; 92:JVI.01270-18. [PMID: 30258000 DOI: 10.1128/jvi.01270-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/14/2018] [Indexed: 12/24/2022] Open
Abstract
Rift Valley fever virus (RVFV) is an arbovirus that causes disease in livestock and humans in Africa and the Middle East. While human disease is typically mild and self-limiting, some individuals develop severe manifestations, such as hepatitis, hemorrhagic fever, or encephalitis. Encephalitis occurs 2 to 3 weeks after acute illness; therefore, we hypothesized that it was a result of an inadequate adaptive immunity. To test this hypothesis in vivo, we used an attenuated virus (DelNSsRVFV) that does not typically cause disease in mice. We first characterized the normal immune response to infection with DelNSsRVFV in immunocompetent mice and noted expansion of natural killer cells and monocytes, as well as activation of both CD8 and CD4 T cells. Depleting C57BL/6 mice of CD4 T cells prior to DelNSsRVFV infection resulted in encephalitis in 30% of the mice; in encephalitic mice, we noted infiltration of T cells and inflammatory monocytes into the brain. CD4 and CD8 codepletion in C57BL/6 mice, as well as CD4 depletion in CCR2 knockout mice, increased the frequency of encephalitis, demonstrating that these cell types normally contributed to the prevention of disease. Encephalitic mice had similar viral RNA loads in the brain regardless of which cell types were depleted, suggesting that CD4 T cells, CD8 T cells, and inflammatory monocytes did little to control viral replication in the brain. CD4-depleted mice exhibited diminished humoral and T cell memory responses, suggesting that these immune mechanisms contributed to peripheral control of virus, thus preventing infection of the brain.IMPORTANCE RVFV is found in Africa and the Middle East and is transmitted by mosquitos or through contact with infected animals. Infected individuals can develop mild disease or more severe forms, such as hepatitis or encephalitis. In order to understand why some individuals develop encephalitis, we first need to know which immune functions protect those who do not develop this form of disease. In this study, we used a mouse model of RVFV infection to demonstrate that CD4 T cells, CD8 T cells, and monocytes all contribute to prevention of encephalitis. Their likely mechanism of action is preventing RVFV from ever reaching the brain.
Collapse
|
29
|
Li G, Teleki C, Wang T. Memory T Cells in Flavivirus Vaccination. Vaccines (Basel) 2018; 6:E73. [PMID: 30340377 PMCID: PMC6313919 DOI: 10.3390/vaccines6040073] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 12/30/2022] Open
Abstract
Flaviviruses include many medically important viruses, such as Dengue virus (DENV), Japanese encephalitis (JEV), tick-borne encephalitis (TBEV), West Nile (WNV), yellow fever (YFV), and Zika viruses (ZIKV). Currently, there are licensed human vaccines for DENV, JEV, TBEV and YFV, but not for WNV or ZIKV. Memory T cells play a central role in adaptive immunity and are important for host protection during flavivirus infection. In this review, we discuss recent findings from animal models and clinical trials and provide new insights into the role of memory T cells in host protective immunity upon vaccination with the licensed flavivirus vaccines.
Collapse
Affiliation(s)
- Guangyu Li
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Cody Teleki
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
30
|
Barzon L, Palù G. Recent developments in vaccines and biological therapies against Japanese encephalitis virus. Expert Opin Biol Ther 2018; 18:851-864. [DOI: 10.1080/14712598.2018.1499721] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Luisa Barzon
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, Padova, Italy
| |
Collapse
|
31
|
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]
|
32
|
Zia A, Singh D, Saxena S, Umrao J, Baluni M, Ghildiyal S, Fatima T, Shukla M, Agarwal V, Dhole TN. Detection of long term cellular immune response to Japanese encephalitis vaccination using IFN-γ ELIspot assay. J Med Virol 2017; 89:2235-2238. [PMID: 28671301 DOI: 10.1002/jmv.24893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/15/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Amreen Zia
- Department of Microbiology; Sanjay Gandhi Post Graduate Institute of Medical Sciences; Lucknow Uttar Pradesh India
| | - Dharamveer Singh
- Department of Microbiology; Sanjay Gandhi Post Graduate Institute of Medical Sciences; Lucknow Uttar Pradesh India
| | - Swati Saxena
- Department of Microbiology; Sanjay Gandhi Post Graduate Institute of Medical Sciences; Lucknow Uttar Pradesh India
| | - Jyoti Umrao
- Department of Microbiology; Sanjay Gandhi Post Graduate Institute of Medical Sciences; Lucknow Uttar Pradesh India
| | - Manjari Baluni
- Department of Microbiology; Sanjay Gandhi Post Graduate Institute of Medical Sciences; Lucknow Uttar Pradesh India
| | - Sneha Ghildiyal
- Department of Microbiology; Sanjay Gandhi Post Graduate Institute of Medical Sciences; Lucknow Uttar Pradesh India
| | - Tanzeem Fatima
- Department of Microbiology; Sanjay Gandhi Post Graduate Institute of Medical Sciences; Lucknow Uttar Pradesh India
| | - Mukti Shukla
- Department of Microbiology; Sanjay Gandhi Post Graduate Institute of Medical Sciences; Lucknow Uttar Pradesh India
| | - Vikas Agarwal
- Department of Clinical Immunology; Sanjay Gandhi Post Graduate Institute of Medical Sciences; Lucknow Uttar Pradesh India
| | - Tapan N. Dhole
- Department of Microbiology; Sanjay Gandhi Post Graduate Institute of Medical Sciences; Lucknow Uttar Pradesh India
| |
Collapse
|
33
|
T Cell Immunity and Zika Virus Vaccine Development. Trends Immunol 2017; 38:594-605. [DOI: 10.1016/j.it.2017.05.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 12/30/2022]
|
34
|
Both haemagglutinin-specific antibody and T cell responses induced by a chimpanzee adenoviral vaccine confer protection against influenza H7N9 viral challenge. Sci Rep 2017; 7:1854. [PMID: 28500340 PMCID: PMC5431854 DOI: 10.1038/s41598-017-02019-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 04/03/2017] [Indexed: 02/06/2023] Open
Abstract
Since 2013, the outbreak or sporadic infection of a new reassortant H7N9 influenza virus in China has resulted in hundreds of deaths and thousands of illnesses. An H7N9 vaccine is urgently needed, as a licensed human vaccine against H7N9 influenza is currently not available. Here, we developed a recombinant adenovirus-based vaccine, AdC68-H7HA, by cloning the H7N9 haemagglutinin (HA) gene into the chimpanzee adenoviral vector AdC68. The efficacy of AdC68-H7HA was evaluated in mice as well as guinea pigs. For comparison, an H7N9 DNA vaccine based on HA was also generated and tested in mice and guinea pigs. The results demonstrated that both AdC68-H7HA and the DNA vaccine prime-adenovirus boost regimen induced potent immune responses in animals and completely protected mice from lethal H7N9 influenza viral challenge. A post-immunization serum transfer experiment showed that antibody responses could completely protect against lethal challenge, while a T cell depletion experiment indicated that HA-specific CD8+ T cells responses also contributed to protection. Therefore, both HA-specific humoral immunity and cellular immunity play important roles in the protection. These data suggest that the chimpanzee adenovirus expressing HA is a promising vaccine candidate for H7N9 virus or other influenza viral subtypes.
Collapse
|
35
|
Partial dysfunction of STAT1 profoundly reduces host resistance to flaviviral infection. Virology 2017; 506:1-6. [PMID: 28282567 DOI: 10.1016/j.virol.2017.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/01/2017] [Accepted: 03/03/2017] [Indexed: 11/22/2022]
Abstract
The genetic basis for a dramatically increased virus susceptibility phenotype of MHC-II knockout mice acquired during routine maintenance of the mouse strain was determined. Segregation of the susceptibility allele from the defective MHC-II locus combined with sequence capture and sequencing showed that a Y37L substitution in STAT1 accounted for high flavivirus susceptibility of a newly derived mouse strain, designated Tuara. Interestingly, the mutation in STAT1 gene gave only partial inactivation of the type I interferon antiviral pathway. Accordingly, merely a relatively small impairment of interferon α/β signalling is sufficient to overcome the ability of the host to control the infection.
Collapse
|
36
|
CD8 T cells protect adult naive mice from JEV-induced morbidity via lytic function. PLoS Negl Trop Dis 2017; 11:e0005329. [PMID: 28151989 PMCID: PMC5308832 DOI: 10.1371/journal.pntd.0005329] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/14/2017] [Accepted: 01/13/2017] [Indexed: 12/02/2022] Open
Abstract
Following Japanese encephalitis virus (JEV) infection neutralizing antibodies are shown to provide protection in a significant proportion of cases, but not all, suggesting additional components of immune system might also contribute to elicit protective immune response. Here we have characterized the role of T cells in offering protection in adult mice infected with JEV. Mice lacking α/β–T cells (TCRβ–null) are highly susceptible and die over 10–18 day period as compared to the wild-type (WT) mice which are resistant. This is associated with high viral load, higher mRNA levels of proinflammatory cytokines and breach in the blood-brain-barrier (BBB). Infected WT mice do not show a breach in BBB; however, in contrast to TCRβ-null, they show the presence of T cells in the brain. Using adoptive transfer of cells with specific genetic deficiencies we see that neither the presence of CD4 T cells nor cytokines such as IL-4, IL-10 or interferon-gamma have any significant role in offering protection from primary infection. In contrast, we show that CD8 T cell deficiency is more critical as absence of CD8 T cells alone increases mortality in mice infected with JEV. Further, transfer of T cells from beige mice with defects in granular lytic function into TCRβ-null mice shows poor protection implicating granule-mediated target cell lysis as an essential component for survival. In addition, for the first time we report that γ/δ-T cells also make significant contribution to confer protection from JEV infection. Our data show that effector CD8 T cells play a protective role during primary infection possibly by preventing the breach in BBB and neuronal damage. Japanese encephalitis virus (JEV) commonly infects human beings in developing countries including those in Southeast Asia. While the majority of the infected people suffer from mild illness, a minority suffers from encephalitis which may lead to death. The virus is transmitted by mosquito bites and elimination of mosquitoes is not a practical answer to prevent the disease, therefore, prevention by vaccination is a desired goal. While various vaccines are clinically tried and some are marketed further improvement in vaccines is still possible. In a complex disease like JE many components of the immune system contribute to variable extent in protection. We show here that one subset of T cells called CD8 cells which are capable of killing infected cells are very critical for providing protection against JEV infection in mice. In the absence of T cells we also observed that virus reaches the brain early, unlike in the presence of T cells, and this possibly results in high virus load in the brain leading to worsening of the condition and death. Thus, our data help in identifying the role of CD8 T cells in protection from lethal JEV infection and the information may be useful for modifying and/or developing vaccine for prevention of JEV-mediated disease.
Collapse
|
37
|
Blom K, Braun M, Pakalniene J, Lunemann S, Enqvist M, Dailidyte L, Schaffer M, Lindquist L, Mickiene A, Michaëlsson J, Ljunggren HG, Gredmark-Russ S. NK Cell Responses to Human Tick-Borne Encephalitis Virus Infection. THE JOURNAL OF IMMUNOLOGY 2016; 197:2762-71. [DOI: 10.4049/jimmunol.1600950] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 07/19/2016] [Indexed: 12/24/2022]
|
38
|
Kim JH, Patil AM, Choi JY, Kim SB, Uyangaa E, Hossain FMA, Park SY, Lee JH, Eo SK. CCR5 ameliorates Japanese encephalitis via dictating the equilibrium of regulatory CD4(+)Foxp3(+) T and IL-17(+)CD4(+) Th17 cells. J Neuroinflammation 2016; 13:223. [PMID: 27439902 PMCID: PMC5050958 DOI: 10.1186/s12974-016-0656-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 07/10/2016] [Indexed: 12/30/2022] Open
Abstract
Background CCR5 is a CC chemokine receptor involved in the migration of effector leukocytes including macrophages, NK, and T cells into inflamed tissues. Also, the role of CCR5 in CD4+Foxp3+ regulatory T cell (Treg) homing has recently begun to grab attention. Japanese encephalitis (JE) is defined as severe neuroinflammation of the central nervous system (CNS) following infection with mosquito-borne flavivirus JE virus. However, the potential contribution of CCR5 to JE progression via mediating CD4+Foxp3+ Treg homing has not been investigated. Methods Infected wild-type (Ccr5+/+) and CCR5-deficient (Ccr5−/−) mice were examined daily for mortality and clinical signs, and neuroinflammation in the CNS was evaluated by infiltration of inflammatory leukocytes and cytokine expression. In addition, viral burden, NK- and JEV-specific T cell responses were analyzed. Adoptive transfer of CCR5+CD4+Foxp3+ Tregs was used to evaluate the role of Tregs in JE progression. Results CCR5 ablation exacerbated JE without altering viral burden in the extraneural and CNS tissues, as manifested by increased CNS infiltration of Ly-6Chi monocytes and Ly-6Ghi granulocytes. Compared to Ccr5+/+ mice, Ccr5−/− mice unexpectedly showed increased responses of IFN-γ+NK and CD8+ T cells in the spleen, but not CD4+ T cells. More interestingly, CCR5-ablation resulted in a skewed response to IL-17+CD4+ Th17 cells and correspondingly reduced CD4+Foxp3+ Tregs in the spleen and brain, which was closely associated with exacerbated JE. Our results also revealed that adoptive transfer of sorted CCR5+CD4+Foxp3+ Tregs into Ccr5−/− mice could ameliorate JE progression without apparently altering the viral burden and CNS infiltration of IL-17+CD4+ Th17 cells, myeloid-derived Ly-6Chi monocytes and Ly-6Ghi granulocytes. Instead, adoptive transfer of CCR5+CD4+Foxp3+ Tregs into Ccr5−/− mice resulted in increased expression of anti-inflammatory cytokines (IL-10 and TGF-β) in the spleen and brain, and transferred CCR5+ Tregs were found to produce IL-10. Conclusions CCR5 regulates JE progression via governing timely and appropriate CNS infiltration of CD4+Foxp3+ Tregs, thereby facilitating host survival. Therefore, this critical and extended role of CCR5 in JE raises possible safety concerns regarding the use of CCR5 antagonists in human immunodeficiency virus (HIV)-infected individuals who inhabit regions in which both HIV and flaviviruses, such as JEV and West Nile virus, are endemic.
Collapse
Affiliation(s)
- Jin Hyoung Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Ajit Mahadev Patil
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Seong Bum Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Erdenebelig Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Ferdaus Mohd Altaf Hossain
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Sang-Youel Park
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea.,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, 54896, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea.,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, 54896, Republic of Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea. .,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, 54896, Republic of Korea.
| |
Collapse
|
39
|
Kim JH, Patil AM, Choi JY, Kim SB, Uyangaa E, Hossain FMA, Park SY, Lee JH, Kim K, Eo SK. CCL2, but not its receptor, is essential to restrict immune privileged central nervous system-invasion of Japanese encephalitis virus via regulating accumulation of CD11b(+) Ly-6C(hi) monocytes. Immunology 2016; 149:186-203. [PMID: 27260136 PMCID: PMC5011677 DOI: 10.1111/imm.12626] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/26/2016] [Accepted: 06/02/2016] [Indexed: 12/15/2022] Open
Abstract
Japanese encephalitis virus (JEV) is a re‐emerging zoonotic flavivirus that poses an increasing threat to global health and welfare due to rapid changes in climate and demography. Although the CCR2–CCL2 axis plays an important role in trafficking CD11b+ Ly‐6Chi monocytes to regulate immunopathological diseases, little is known about their role in monocyte trafficking during viral encephalitis caused by JEV infection. Here, we explored the role of CCR2 and its ligand CCL2 in JE caused by JEV infection using CCR2‐ and CCL2‐ablated murine models. Somewhat surprisingly, the ablation of CCR2 and CCL2 resulted in starkly contrasting susceptibility to JE. CCR2 ablation induced enhanced resistance to JE, whereas CCL2 ablation highly increased susceptibility to JE. This contrasting regulation of JE progression by CCR2 and CCL2 was coupled to central nervous system (CNS) infiltration of Ly‐6Chi monocytes and Ly‐6Ghi granulocytes. There was also enhanced expression of CC and CXC chemokines in the CNS of CCL2‐ablated mice, which appeared to induce CNS infiltration of these cell populations. However, our data revealed that contrasting regulation of JE in CCR2‐ and CCL2‐ablated mice was unlikely to be mediated by innate natural killer and adaptive T‐cell responses. Furthermore, CCL2 produced by haematopoietic stem cell‐derived leucocytes played a dominant role in CNS accumulation of Ly‐6Chi monocytes in infected bone marrow chimeric models, thereby exacerbating JE progression. Collectively, our data indicate that CCL2 plays an essential role in conferring protection against JE caused by JEV infection. In addition, blockage of CCR2, but not CCL2, will aid in the development of strategies for prophylactics and therapeutics of JE.
Collapse
Affiliation(s)
- Jin Hyoung Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Korea
| | - Ajit Mahadev Patil
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Korea
| | - Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Korea
| | - Seong Bum Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Korea
| | - Erdenebileg Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Korea
| | - Ferdaus Mohd Altaf Hossain
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Korea
| | - Sang-Youel Park
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Korea.,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, Korea
| | - John Hwa Lee
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Korea.,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, Korea.,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, Korea
| |
Collapse
|
40
|
Turtle L, Bali T, Buxton G, Chib S, Chan S, Soni M, Hussain M, Isenman H, Fadnis P, Venkataswamy MM, Satishkumar V, Lewthwaite P, Kurioka A, Krishna S, Shankar MV, Ahmed R, Begum A, Ravi V, Desai A, Yoksan S, Fernandez S, Willberg CB, Kloverpris HN, Conlon C, Klenerman P, Satchidanandam V, Solomon T. Human T cell responses to Japanese encephalitis virus in health and disease. J Exp Med 2016; 213:1331-52. [PMID: 27242166 PMCID: PMC4925015 DOI: 10.1084/jem.20151517] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 05/04/2016] [Indexed: 12/28/2022] Open
Abstract
Japanese encephalitis (JE) virus (JEV) is an important cause of encephalitis in children of South and Southeast Asia. However, the majority of individuals exposed to JEV only develop mild symptoms associated with long-lasting adaptive immunity. The related flavivirus dengue virus (DENV) cocirculates in many JEV-endemic areas, and clinical data suggest cross-protection between DENV and JEV. To address the role of T cell responses in protection against JEV, we conducted the first full-breadth analysis of the human memory T cell response using a synthetic peptide library. Ex vivo interferon-γ (IFN-γ) responses to JEV in healthy JEV-exposed donors were mostly CD8(+) and targeted nonstructural (NS) proteins, whereas IFN-γ responses in recovered JE patients were mostly CD4(+) and targeted structural proteins and the secreted protein NS1. Among patients, a high quality, polyfunctional CD4(+) T cell response was associated with complete recovery from JE. T cell responses from healthy donors showed a high degree of cross-reactivity to DENV that was less apparent in recovered JE patients despite equal exposure. These data reveal divergent functional CD4(+) and CD8(+) T cell responses linked to different clinical outcomes of JEV infection, associated with distinct targeting and broad flavivirus cross-reactivity including epitopes from DENV, West Nile, and Zika virus.
Collapse
Affiliation(s)
- Lance Turtle
- Institute of Infection and Global Health, University of Liverpool, Liverpool L69 7BE, England, UK
- Health Protection Research Unit for Emerging and Zoonotic Infections, University of Liverpool, Liverpool L69 7BE, England, UK
- Tropical and Infectious Disease Unit, Royal Liverpool University Hospital, Liverpool L7 8XP, England, UK
| | - Tanushka Bali
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Gemma Buxton
- Institute of Infection and Global Health, University of Liverpool, Liverpool L69 7BE, England, UK
| | - Savita Chib
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Sajesh Chan
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Mohammed Soni
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Mohammed Hussain
- Department of Microbiology, Vijayanagar Institute of Medical Science Medical College, Bellary 583104, India
| | - Heather Isenman
- Institute of Infection and Global Health, University of Liverpool, Liverpool L69 7BE, England, UK
| | - Prachi Fadnis
- Department of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Manjunatha M. Venkataswamy
- Department of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Vishali Satishkumar
- Department of Microbiology, Vijayanagar Institute of Medical Science Medical College, Bellary 583104, India
- Department of Paediatrics, Vijayanagar Institute of Medical Science Medical College, Bellary 583104, India
| | - Penny Lewthwaite
- Department of Infection and Travel Medicine, University Hospital of St. James, Leeds Teaching Hospitals, National Health Service Trust, Leeds LS9 7TF, England, UK
| | - Ayako Kurioka
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford OX1 3SY, England, UK
| | - Srinivasa Krishna
- Department of Microbiology, Vijayanagar Institute of Medical Science Medical College, Bellary 583104, India
| | - M. Veera Shankar
- Department of Paediatrics, Vijayanagar Institute of Medical Science Medical College, Bellary 583104, India
| | - Riyaz Ahmed
- Department of Paediatrics, Vijayanagar Institute of Medical Science Medical College, Bellary 583104, India
| | - Ashia Begum
- Department of Microbiology, Vijayanagar Institute of Medical Science Medical College, Bellary 583104, India
- Department of Paediatrics, Vijayanagar Institute of Medical Science Medical College, Bellary 583104, India
| | - Vasanthapuram Ravi
- Department of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Anita Desai
- Department of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Sutee Yoksan
- Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University at Salaya, Bangkok 73170, Thailand
| | - Stefan Fernandez
- Department of Virology, Armed Forces Research Institute of Medical Science, Bangkok 10400, Thailand
| | - Christian B. Willberg
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford OX1 3SY, England, UK
| | - Henrik N. Kloverpris
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford OX1 3SY, England, UK
| | - Christopher Conlon
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford OX1 3SY, England, UK
| | - Paul Klenerman
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford OX1 3SY, England, UK
| | - Vijaya Satchidanandam
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Tom Solomon
- Institute of Infection and Global Health, University of Liverpool, Liverpool L69 7BE, England, UK
- Health Protection Research Unit for Emerging and Zoonotic Infections, University of Liverpool, Liverpool L69 7BE, England, UK
- Walton Center National Health Service Foundation Trust, Liverpool L9 7LJ, England, UK
| |
Collapse
|
41
|
Kim SB, Choi JY, Uyangaa E, Patil AM, Hossain FMA, Hur J, Park SY, Lee JH, Kim K, Eo SK. Blockage of indoleamine 2,3-dioxygenase regulates Japanese encephalitis via enhancement of type I/II IFN innate and adaptive T-cell responses. J Neuroinflammation 2016; 13:79. [PMID: 27090635 PMCID: PMC4835894 DOI: 10.1186/s12974-016-0551-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/12/2016] [Indexed: 11/24/2022] Open
Abstract
Background Japanese encephalitis (JE), a leading cause of viral encephalitis, is characterized by extensive neuroinflammation following infection with neurotropic JE virus (JEV). Indoleamine 2,3-dioxygenase (IDO) has been identified as an enzyme associated with immunoregulatory function. Although the regulatory role of IDO in viral replication has been postulated, the in vivo role of IDO activity has not been fully addressed in neurotropic virus-caused encephalitis. Methods Mice in which IDO activity was inhibited by genetic ablation or using a specific inhibitor were examined for mortality and clinical signs after infection. Neuroinflammation was evaluated by central nervous system (CNS) infiltration of leukocytes and cytokine expression. IDO expression, viral burden, JEV-specific T-cell, and type I/II interferon (IFN-I/II) innate responses were also analyzed. Results Elevated expression of IDO activity in myeloid and neuron cells of the lymphoid and CNS tissues was closely associated with clinical signs of JE. Furthermore, inhibition of IDO activity enhanced resistance to JE, reduced the viral burden in lymphoid and CNS tissues, and resulted in early and increased CNS infiltration by Ly-6Chi monocytes, NK, CD4+, and CD8+ T-cells. JE amelioration in IDO-ablated mice was also associated with enhanced NK and JEV-specific T-cell responses. More interestingly, IDO ablation induced rapid enhancement of type I IFN (IFN-I) innate responses in CD11c+ dendritic cells (DCs), including conventional and plasmacytoid DCs, following JEV infection. This enhanced IFN-I innate response in IDO-ablated CD11c+ DCs was coupled with strong induction of PRRs (RIG-I, MDA5), transcription factors (IRF7, STAT1), and antiviral ISG genes (Mx1, Mx2, ISG49, ISG54, ISG56). IDO ablation also enhanced the IFN-I innate response in neuron cells, which may delay the spread of virus in the CNS. Finally, we identified that IDO ablation in myeloid cells derived from hematopoietic stem cells (HSCs) dominantly contributed to JE amelioration and that HSC-derived leukocytes played a key role in the enhanced IFN-I innate responses in the IDO-ablated environment. Conclusions Inhibition of IDO activity ameliorated JE via enhancement of antiviral IFN-I/II innate and adaptive T-cell responses and increased CNS infiltration of peripheral leukocytes. Therefore, our data provide valuable insight into the use of IDO inhibition by specific inhibitors as a promising tool for therapeutic and prophylactic strategies against viral encephalitis caused by neurotropic viruses.
Collapse
Affiliation(s)
- Seong Bum Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Erdenebileg Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Ajit Mahadev Patil
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Ferdaus Mohd Altaf Hossain
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Jin Hur
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Sang-Youel Park
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea.,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, 54896, Republic of Korea
| | - John-Hwa Lee
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea.,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, 54896, Republic of Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea. .,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, 54896, Republic of Korea.
| |
Collapse
|
42
|
Abstract
Dengue provides the most abundant example in human medicine and the greatest human illness burden caused by the phenomenon of intrinsic antibody-dependent infection enhancement (iADE). In this immunopathological phenomenon infection of monocytes or macrophages using infectious immune complexes suppresses innate antiviral systems, permitting logarithmic intracellular growth of dengue virus. The four dengue viruses evolved from a common ancestor yet retain similar ecology and pathogenicity, but although infection with one virus provides short-term cross-protection against infection with a different type, millions of secondary dengue infections occur worldwide each year. When individuals are infected in the virtual absence of cross-protective dengue antibodies, the dengue vascular permeability syndrome (DVPS) may ensue. This occurs in around 2 to 4% of second heterotypic dengue infections. A complete understanding of the biologic mechanism of iADE, dengue biology, and the mechanism of host responses to dengue infection should lead to a comprehensive and complete understanding of the pathogenesis of DVPS. A crucial emphasis must be placed on understanding ADE. Clinical and epidemiological observations of DVPS define the research questions and provide research parameters. This article will review knowledge related to dengue ADE and point to areas where there has been little research progress. These observations relate to the two stages of dengue illnesses: afferent phenomena are those that promote the success of the microorganism to infect and survive; efferent phenomena are those mounted by the host to inhibit infection and replication and to eliminate the infectious agent and infected tissues. Data will be discussed as "knowns" and "unknowns."
Collapse
|
43
|
A Single Amino Acid Substitution in the M Protein Attenuates Japanese Encephalitis Virus in Mammalian Hosts. J Virol 2015; 90:2676-89. [PMID: 26656690 DOI: 10.1128/jvi.01176-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 11/30/2015] [Indexed: 02/04/2023] Open
Abstract
UNLABELLED Japanese encephalitis virus (JEV) membrane (M) protein plays important structural roles in the processes of fusion and maturation of progeny virus during cellular infection. The M protein is anchored in the viral membrane, and its ectodomain is composed of a flexible N-terminal loop and a perimembrane helix. In this study, we performed site-directed mutagenesis on residue 36 of JEV M protein and showed that the resulting mutation had little or no effect on the entry process but greatly affected virus assembly in mammalian cells. Interestingly, this mutant virus had a host-dependent phenotype and could develop a wild-type infection in insect cells. Experiments performed on infectious virus as well as in a virus-like particle (VLP) system indicate that the JEV mutant expresses structural proteins but fails to form infectious particles in mammalian cells. Using a mouse model for JEV pathogenesis, we showed that the mutation conferred complete attenuation in vivo. The production of JEV neutralizing antibodies in challenged mice was indicative of the immunogenicity of the mutant virus in vivo. Together, our results indicate that the introduction of a single mutation in the M protein, while being tolerated in insect cells, strongly impacts JEV infection in mammalian hosts. IMPORTANCE JEV is a mosquito-transmitted flavivirus and is a medically important pathogen in Asia. The M protein is thought to be important for accommodating the structural rearrangements undergone by the virion during viral assembly and may play additional roles in the JEV infectious cycle. In the present study, we show that a sole mutation in the M protein impairs the JEV infection cycle in mammalian hosts but not in mosquito cells. This finding highlights differences in flavivirus assembly pathways among hosts. Moreover, infection of mice indicated that the mutant was completely attenuated and triggered a strong immune response to JEV, thus providing new insights for further development of JEV vaccines.
Collapse
|
44
|
Kim SB, Choi JY, Kim JH, Uyangaa E, Patil AM, Park SY, Lee JH, Kim K, Han YW, Eo SK. Amelioration of Japanese encephalitis by blockage of 4-1BB signaling is coupled to divergent enhancement of type I/II IFN responses and Ly-6C(hi) monocyte differentiation. J Neuroinflammation 2015; 12:216. [PMID: 26597582 PMCID: PMC4657197 DOI: 10.1186/s12974-015-0438-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/16/2015] [Indexed: 12/28/2022] Open
Abstract
Background Japanese encephalitis (JE), a neuroinflammation caused by zoonotic JE virus, is the major cause of viral encephalitis worldwide and poses an increasing threat to global health and welfare. To date, however, there has been no report describing the regulation of JE progression using immunomodulatory tools for developing therapeutic strategies. We tested whether blocking the 4-1BB signaling pathway would regulate JE progression using murine JE model. Methods Infected wild-type and 4-1BB-knockout (KO) mice were examined daily for mortality and clinical signs, and neuroinflammation in the CNS was evaluated by infiltration of inflammatory leukocytes and cytokine expression. In addition, viral burden, JEV-specific T cell, and type I/II IFN (IFN-I/II) innate responses were analyzed. Results Blocking the 4-1BB signaling pathway significantly increased resistance to JE and reduced viral burden in extraneural tissues and the CNS, rather than causing a detrimental effect. In addition, treatment with 4-1BB agonistic antibody exacerbated JE. Furthermore, JE amelioration and reduction of viral burden by blocking the 4-1BB signaling pathway were associated with an increased frequency of IFN-II-producing NK and CD4+ Th1 cells as well as increased infiltration of mature Ly-6Chi monocytes in the inflamed CNS. More interestingly, DCs and macrophages derived from 4-1BB KO mice showed potent and rapid IFN-I innate immune responses upon JEV infection, which was coupled to strong induction of PRRs (RIG-I, MDA5), transcription factors (IRF7), and antiviral ISG genes (ISG49, ISG54, ISG56). Further, the ablation of 4-1BB signaling enhanced IFN-I innate responses in neuron cells, which likely regulated viral spread in the CNS. Finally, we confirmed that blocking the 4-1BB signaling pathway in myeloid cells derived from hematopoietic stem cells (HSCs) played a dominant role in ameliorating JE. In support of this finding, HSC-derived leukocytes played a dominant role in generating the IFN-I innate responses in the host. Conclusions Blocking the 4-1BB signaling pathway ameliorates JE via divergent enhancement of IFN-II-producing NK and CD4+ Th1 cells and mature Ly-6Chi monocyte infiltration, as well as an IFN-I innate response of myeloid-derived cells. Therefore, regulation of the 4-1BB signaling pathway with antibodies or inhibitors could be a valuable therapeutic strategy for the treatment of JE.
Collapse
Affiliation(s)
- Seong Bum Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Jin Hyoung Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Erdenebelig Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Ajit Mahadev Patil
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Sang-Youel Park
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea.,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, 54896, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea.,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, 54896, Republic of Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Young Woo Han
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan, 54596, Republic of Korea. .,Department of Bioactive Material Sciences, Graduate School, Chonbuk National University, Jeonju, 54896, Republic of Korea.
| |
Collapse
|
45
|
Gupta N, de Wispelaere M, Lecerf M, Kalia M, Scheel T, Vrati S, Berek C, Kaveri SV, Desprès P, Lacroix-Desmazes S, Dimitrov JD. Neutralization of Japanese Encephalitis Virus by heme-induced broadly reactive human monoclonal antibody. Sci Rep 2015; 5:16248. [PMID: 26542535 PMCID: PMC4635365 DOI: 10.1038/srep16248] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 10/12/2015] [Indexed: 12/04/2022] Open
Abstract
Geographical expansion and re-emerging new genotypes of the Japanese encephalitis virus (JEV) require the development of novel therapeutic approaches. Here, we studied a non-conventional approach for antibody therapy and show that, upon exposure to heme, a fraction of natural human immunoglobulins acquires high-affinity reactivity with the antigenic domain-III of JEV E glycoprotein. These JEV-reactive antibodies exhibited neutralizing activity against recently dominant JEV genotypes. This study opens new therapeutic options for Japanese encephalitis.
Collapse
Affiliation(s)
- Nimesh Gupta
- INSERM, UMR S 1138, Centre de Recherche des Cordeliers, Paris, France.,Université Pierre et Marie Curie-Paris6, UMR S 1138, Paris, France.,Université Paris Descartes, UMR S 1138, Paris, France
| | - Mélissanne de Wispelaere
- Flavivirus-Host Molecular Interactions laboratory, Virology Department, Institut Pasteur, Paris, France
| | - Maxime Lecerf
- INSERM, UMR S 1138, Centre de Recherche des Cordeliers, Paris, France.,Université Pierre et Marie Curie-Paris6, UMR S 1138, Paris, France.,Université Paris Descartes, UMR S 1138, Paris, France
| | - Manjula Kalia
- Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Tobias Scheel
- Deutsches Rheuma-Forschungszentrum, Institut der Leibniz-Gemeinschaft, 10117 Berlin, Germany
| | - Sudhanshu Vrati
- Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Claudia Berek
- Deutsches Rheuma-Forschungszentrum, Institut der Leibniz-Gemeinschaft, 10117 Berlin, Germany
| | - Srinivas V Kaveri
- INSERM, UMR S 1138, Centre de Recherche des Cordeliers, Paris, France.,Université Pierre et Marie Curie-Paris6, UMR S 1138, Paris, France.,Université Paris Descartes, UMR S 1138, Paris, France
| | - Philippe Desprès
- Flavivirus-Host Molecular Interactions laboratory, Virology Department, Institut Pasteur, Paris, France.,Infection and Epidemiology Department, Institut Pasteur, 75724 Paris &UMR PIMIT (I2T), University of Reunion Island, INSERM U1187, CNRS 9192, IRD 249, GIP-CYROI, la Reunion, France
| | - Sébastien Lacroix-Desmazes
- INSERM, UMR S 1138, Centre de Recherche des Cordeliers, Paris, France.,Université Pierre et Marie Curie-Paris6, UMR S 1138, Paris, France.,Université Paris Descartes, UMR S 1138, Paris, France
| | - Jordan D Dimitrov
- INSERM, UMR S 1138, Centre de Recherche des Cordeliers, Paris, France.,Université Pierre et Marie Curie-Paris6, UMR S 1138, Paris, France.,Université Paris Descartes, UMR S 1138, Paris, France
| |
Collapse
|
46
|
A Japanese encephalitis virus genotype 5 molecular clone is highly neuropathogenic in a mouse model: impact of the structural protein region on virulence. J Virol 2015; 89:5862-75. [PMID: 25787283 DOI: 10.1128/jvi.00358-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/11/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Japanese encephalitis virus (JEV) strains can be separated into 5 genotypes (g1 to g5) based on sequence similarity. JEV g5 strains have been rarely isolated and are poorly characterized. We report here the full characterization of a g5 virus generated using a cDNA-based technology and its comparison with a widely studied g3 strain. We did not observe any major differences between those viruses when their infectious cycles were studied in various cell lines in vitro. Interestingly, the JEV g5 strain was highly pathogenic when inoculated to BALB/c mice, which are known to be largely resistant to JEV g3 infection. The study of chimeric viruses between JEV g3 and g5 showed that there was a poor viral clearance of viruses that express JEV g5 structural proteins in BALB/c mice blood, which correlated with viral invasion of the central nervous system and encephalitis. In addition, using an in vitro model of the blood-brain barrier, we were able to show that JEV g5 does not have an enhanced capacity for entering the central nervous system, compared to JEV g3. Overall, in addition to providing a first characterization of the understudied JEV g5, our work highlights the importance of sustaining an early viremia in the development of JEV encephalitis. IMPORTANCE Genotype 5 viruses are genetically and serologically distinct from other JEV genotypes and can been associated with human encephalitis, which warrants the need for their characterization. In this study, we characterized the in vitro and in vivo properties of a JEV g5 strain and showed that it was more neuropathogenic in a mouse model than a well-characterized JEV g3 strain. The enhanced virulence of JEV g5 was associated with poor viral clearance but not with enhanced crossing of the blood-brain barrier, thus providing new insights into JEV pathogenesis.
Collapse
|
47
|
Swanson PA, McGavern DB. Viral diseases of the central nervous system. Curr Opin Virol 2015; 11:44-54. [PMID: 25681709 DOI: 10.1016/j.coviro.2014.12.009] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/17/2014] [Indexed: 11/18/2022]
Abstract
Virus-induced diseases of the central nervous system (CNS) represent a significant burden to human health worldwide. The complexity of these diseases is influenced by the sheer number of different neurotropic viruses, the diverse routes of CNS entry, viral tropism, and the immune system. Using a combination of human pathological data and experimental animal models, we have begun to uncover many of the mechanisms that viruses use to enter the CNS and cause disease. This review highlights a selection of neurotropic viruses that infect the CNS and explores the means by which they induce neurological diseases such as meningitis, encephalitis, and myelitis.
Collapse
Affiliation(s)
- Phillip A Swanson
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, United States
| | - Dorian B McGavern
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
48
|
Bassi MR, Kongsgaard M, Steffensen MA, Fenger C, Rasmussen M, Skjødt K, Finsen B, Stryhn A, Buus S, Christensen JP, Thomsen AR. CD8+ T cells complement antibodies in protecting against yellow fever virus. THE JOURNAL OF IMMUNOLOGY 2014; 194:1141-53. [PMID: 25539816 DOI: 10.4049/jimmunol.1402605] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The attenuated yellow fever (YF) vaccine (YF-17D) was developed in the 1930s, yet little is known about the protective mechanisms underlying its efficiency. In this study, we analyzed the relative contribution of cell-mediated and humoral immunity to the vaccine-induced protection in a murine model of YF-17D infection. Using different strains of knockout mice, we found that CD4(+) T cells, B cells, and Abs are required for full clinical protection of vaccinated mice, whereas CD8(+) T cells are dispensable for long-term survival after intracerebral challenge. However, by analyzing the immune response inside the infected CNS, we observed an accelerated T cell influx into the brain after intracerebral challenge of vaccinated mice, and this T cell recruitment correlated with improved virus control in the brain. Using mice deficient in B cells we found that, in the absence of Abs, YF vaccination can still induce some antiviral protection, and in vivo depletion of CD8(+) T cells from these animals revealed a pivotal role for CD8(+) T cells in controlling virus replication in the absence of a humoral response. Finally, we demonstrated that effector CD8(+) T cells also contribute to viral control in the presence of circulating YF-specific Abs. To our knowledge, this is the first time that YF-specific CD8(+) T cells have been demonstrated to possess antiviral activity in vivo.
Collapse
Affiliation(s)
- Maria R Bassi
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Michael Kongsgaard
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Maria A Steffensen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Christina Fenger
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark; and
| | - Michael Rasmussen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Karsten Skjødt
- Department of Cancer and Inflammation, Institute for Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark
| | - Bente Finsen
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark; and
| | - Anette Stryhn
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Søren Buus
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Jan P Christensen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Allan R Thomsen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen N, Denmark;
| |
Collapse
|
49
|
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
|
50
|
A poorly neutralizing IgG2a/c response elicited by a DNA vaccine protects mice against Japanese encephalitis virus. J Gen Virol 2014; 95:1983-1990. [DOI: 10.1099/vir.0.067280-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
We demonstrated previously that immunization with a DNA vaccine expressing the Japanese encephalitis virus (JEV) envelope (E) protein conferred a high level of protection through a poorly neutralizing antibody response. Here, we further investigated the role of the IgG subclass in this antibody-dependent protection using cytokine co-immunization and cytokine-deficient mice. A significant difference in IgG2a/c but not IgG1 was observed between mice that survived or died following a lethal challenge. Correspondingly, the IgG2a/c response and protection increased in IL-4-deficient mice but decreased in IFN-γ-deficient mice, highlighting the importance of IgG2a/c. In addition, the restoration of protection and E-specific IgG2a/c production in IFN-γ-deficient mice by a T helper (Th) type 1-biased intramuscular immunization suggested that IgG2a/c but not IFN-γ was the major component for protection. The failure of protection against a direct intracranial challenge indicated that IgG2a/c-mediated protection was restricted to outside the central nervous system. Consistent with this conclusion, passive transfer of E-specific antisera conferred protection only pre-exposure to JEV. Therefore, our data provided evidence that the IgG subclass plays an important role in protection against JEV, particular in poorly neutralizing E-specific antibodies, and Th1-biased IgG2a/c confers better protection than Th2-biased IgG1 against JEV.
Collapse
|