Elsevier

Microbial Pathogenesis

Volume 136, November 2019, 103678
Microbial Pathogenesis

Japanese encephalitis virus: Associated immune response and recent progress in vaccine development

https://doi.org/10.1016/j.micpath.2019.103678Get rights and content

Highlights

  • Japanese encephalitis (JE) is the main cause of viral encephalitis worldwide.

  • Majorities of Asian and pacific countries are endemic for the disease in which more than three billion people are at risks of infection.

  • JE virus has complex eco-epidemiology, which limits the effectiveness of vector control strategies.

  • Vaccination is the more effective to prevent the infection where JEV is a major public health problem.

Abstract

Japanese encephalitis (JE) has emerged as one of the most important form of viral encephalitis, which accounts for an estimated 70,000 cases each year with approximately 10,000 fatalities. The clinical presentations and outcome of the infection is dependent upon both virulence of viral determinants and host immune responses. The causative pathogen of JE is a virus known as Japanese encephalitis virus (JEV), which penetrates into the CNS from blood and triggers rapid humoral and cell-mediated immune response. Humoral response is crucial for the control of dissemination of JEV infection and the cytokines produced by cell-mediated immunity during JEV infections serve as potent immune mediators. Till date, JE is only vaccine preventable and no complete antiviral treatment is available so far. Further, vaccine-mediated prevention also has certain limitations. Therefore, an understanding of the pathogenesis of JEV infection can enable the researchers to presume the depth of treatment regime. This review highlights the importance of understanding of the immune mechanisms that are operated in the host during JEV infection and would be helpful in improving future vaccination strategy against JEV.

Introduction

Japanese encephalitis virus (JEV) is a member of genus Flavivirus that belongs to family Flaviviridae. It is an enveloped virus consisting of single stranded positive sense RNA genome surrounded by a nucleocapsid. The virus is transmitted to humans by mosquito vector, principally by Culex tritaeniorhynchus that proliferates in close proximity with other vertebrate hosts like pigs, water birds and chicks. JEV is an etiological agent of acute zoonotic infection, commonly affecting children and is the major cause of epidemic Japanese encephalitis (JE) in Asia [1]. Since 1870s, JE has emerged as the most important form of viral encephalitis throughout South-east Asia, China, and in Asia-pacific belt [2]. It is a serious central nervous system (CNS) disease with high rate of mortality and morbidity, and has a potential to spread to far unaffected areas like Australian continent [3]. JE has been attributed to an estimated worldwide incidence of 50,000 to 70,000 cases with a high case fatality rate of 30–50%. A large proportion of survivors suffer from long-term neurological manifestations in the form of convulsions, tremors, paralysis, ataxia and other such symptoms [2]. Different disease manifestations however occur in the host as a result of JEV infection, ranging from mild subclinical febrile illness where infection is limited to the extraneural tissues to the clinical infections leading to encephalitis.

JEV is transmitted to humans from the bite of carrier mosquito and it replicates in local tissues and regional lymph nodes from where it is carried via lymphatics to the thoracic duct and then into the bloodstream. From blood, the virus penetrates into the CNS and causes encephalitis. A rapid immune response is generated in the organs where JEV replicates with mononuclear and polymorphonuclear cell infiltrations [4]. However, the mechanism involved in producing this inflammatory state is not clearly understood [5] and an elucidation of immune responses to JEV infection has been an active area of research during the past few decades in both humans and experimental animals [[6], [7], [8], [9], [10]]. At present, both humoral [11] and cell-mediated immunity (CMI) have been implicated in providing protection against JEV infection, where early host defense against JEV infection is mediated by phagocytic cells [12], and B and T effector cells [6]. However, the role of immune responses in recovery from JEV infection is poorly understood.

The humoral response is indispensable for the control and dissemination of JEV infection. Much of this control is provided by the neutralizing antibodies, which recognize epitopes located predominately in the viral envelope (E) glycoprotein. These antibodies inhibit viral attachment, internalization, and/or replication within the cells. Apart from anti-E antibodies, anti-non-structural-1 (NS-1) and precursor membrane protein (PrM) antibodies are also important in mediating protective immunity to JEV infection [[13], [14], [15]]. A failure to mount adequate humoral immune response by the host can increase the susceptibility to encephalitis caused by JEV. In such a condition, cellular immunity can play an important protective role against JEV infection; however, it is less well characterized. CD4+ and CD8+ T cells are reported to be important in controlling experimental murine flaviviral infections [6,8,[16], [17], [18]]. Pan et al. (2001) have however reported that CD8+ cytotoxic T cell activity is not required for protection and presence of CD4+ T helper (Th) cells assist in the induction of optimal antibody responses by DNA or live JEV vaccines. CD4+ Th cells exert most of their functions through secreted cytokines [19]. Changes in the pattern of cytokines produced by Th cells can change the type of immune responses that develop among other leukocytes. The Th1 response generates a cytokine profile that supports inflammation and activation of certain T cells and macrophages, whereas Th2 response activates B cells and immune responses that depend on antibodies. The Th1 cells produce proinflammatory cytokines such as IFN-γ, IL-2 and IL-12, while the Th2 cells produce antiinflammatory cytokines that are involved in the regulation of the B cell response such as IL-4, IL-5, IL -6, and IL-10. However, there is dearth of information in JEV infection regarding the type of Th responses (Th1/Th2) generated as well as the role of cytokines involved. In the study where cytokines and IgG subtypes responses have been studied, the Th responses were found to be mice strain and route specific. These studies involved three stains of mice (C57BL/6J, Swiss Albino and BALB/c), which were immunized with either live JEV (Vellore strain P20778) or killed JEV vaccine by intraperitoneal, subcutaneous and peroral routes. The live JEV was found to induce Th1, while killed vaccine induced a predominant Th2 profile [9].

After the viral exposure, T cells activation leads to the expression of large number of cytokines that facilitate in generation of both cell-mediated and antibody responses against foreign antigen. The early innate and cellular immune responses with special reference to cytokines during JEV infection has been less studied, though there is increasing evidence for the critical role of cytokines during JE. Proinflammatory cytokines like IFN-γ, TNF-α, macrophage migration inhibitory factor and chemokine IL-8, have been found to be associated with bad outcome in small studies including mice brain [[20], [21], [22], [23]]. It has also been shown in a large patient based study that elevated levels of proinflammatory cytokines like IL-6 and chemokines like RANTES (regulated upon activation, T cell expressed and secreted) are associated with a poor outcome, but whether these enhanced the levels of proinflammatory cytokines contribute to the immunopathogenesis or are simply correlated to severe illness is still ambiguous [24]. An in vitro study has recently demonstrated that after JEV infection there is microglial activation followed by subsequent release of various proinflammatory mediators, which induces neuronal death. It has also been reported that although initiation of immune responses by microglial cells is an important protective mechanism in the CNS, unrestrained inflammatory responses may result in irreparable brain damage [25].

The mechanisms by which many neurotropic viruses cause the neurological diseases, precise mechanism of inflammation and immune responses are not fully understood. Infiltration of inflammatory cells, perivascular cuffing, gliosis and necrosis are the hallmarks of brain pathology during JE [22,26,27]. In many cases, virus mediates damage to brain tissues indirectly by triggering cell-mediated immune responses like activation of cytotoxic T cells and macrophages, instead of playing a direct role. Activated inflammatory cells secrete various cytokines, such as IL-1, TNF-α, thereby causing toxic effect in the brain [28]. Although, these immune/inflammatory infiltrates are aimed at restricting the viral growth in the brain, their uncontrolled production is detrimental to the host. Most cytokine studies have been focused on the importance of innate immune response in brain against this neurotropic virus in mice model [22,25]. Since the immune responses in the CNS differs from that in the periphery, the importance of immune response with special reference to cytokines cannot be overlooked in the later system. Here in this review, we discuss JEV-associated immune responses and the contribution of cytokines in the immunopathogenesis of JEV during the course of infection. An understanding of immunopathogenesis associated with JEV can aid in improving the strategies for development of future vaccines against JEV.

Section snippets

Japanese encephalitis virus

JEV was first isolated in 1924 from a clinical case during the first reported epidemic of Japan [29]. In 1935, the prototype Nakayama strain was isolated from the brain of a patient suffering from encephalitis. Thereafter the virus was classified with other flavivirus as group B arbovirus in family Togaviridae, but in 1985 it has been designated under a separate family Flaviviridae as a member of genus Flavivirus [30]. The genus Flavivirus has been named after the prototype yellow fever virus

Immune response

In the early phase of the disease, when the virus replicates and spreads by haematogenous route to other parts of the body, the immune responses are likely to determine the outcome of the host-pathogen interactions [56]. A differential clinical presentation of disease symptoms is observed in JEV-infected individuals where only a small proportion of infected individuals develop clinical features, and these may range from a non-specific flu-like illness to a severe fatal meningoencephalitis,

Vaccination strategies for JEV

There are majorly three types of JE vaccines, which are currently in use: mouse-brain derived inactivated, cell-culture derived inactivated and cell-culture derived live attenuated JE vaccine. The formalin-inactivated vaccines are reported to be safe and effective against JEV for at least 30 years [33]. However, the mouse-brain inactivated vaccines were the most widely produced and internationally distributed vaccines, which are no longer in use owing to concerns regarding adverse effects of

Conclusions

The fundamental basis of vaccination is the generation and maintenance of an antigen-specific immune response, which is sufficient to mediate protection from infection and trigger a long-lived humoral response through the sufficient production of antibodies. In order to develop such an effective vaccine against JEV, it is important to understand the mechanisms of the immune response to JEV infection, and we have critically dealt with it in this review.

A variety of approaches have been tested

Conflicts of interest

The authors have no competing interests to declare.

Acknowledgements

The author KKN is the recipient of Ramalingaswami re-entry fellowship and acknowledges the Department of Biotechnology, New Delhi, India. The authors are thankful to the All India Institute of Medical Science, Jodhpur, India and the National Institute of Technology Raipur, India for providing the facility and space.

References (129)

  • G.W. Kreutzberg

    Microglia: a sensor for pathological events in the CNS

    Trends Neurosci.

    (1996)
  • S. Bhowmick et al.

    Induction of IP-10 (CXCL10) in astrocytes following Japanese encephalitis

    Neurosci. Lett.

    (2007)
  • T. Solomon et al.

    Interferon alfa-2a in Japanese encephalitis: a randomized double-blind placebo-controlled trial

    Lancet

    (2003)
  • S.K. Saxena et al.

    Induction of nitric oxide synthase during Japanese encephalitis virus infection: evidence of protective role

    Arch. Biochem. Biophys.

    (2001)
  • D.H. Libraty et al.

    Clinical and immunological risk factors for severe disease in Japanese encephalitis

    Trans. R. Soc. Trop. Med. Hyg.

    (2002)
  • M. Kutubuddin et al.

    Recognition of helper T cell epitopes in envelope (E) glycoprotein of Japanese encephalitis, west Nile and Dengue viruses

    Mol. Immunol.

    (1991)
  • T. Komastu et al.

    IL-12 and viral infections

    Cytokine Growth Factor Rev.

    (1998)
  • T.P. Monath

    Flaviviruses

  • T.F. Tsai et al.

    Japanese encephalitis vaccine

    Vaccine

    (1999)
  • K. Bharati et al.

    Japanese encephalitis: development of new candidate vaccines

    Expert Rev. Anti. Infect. Ther.

    (2006)
  • A. Mathur et al.

    Immunopathological study of spleen during Japanese encephalitis virus infection in mice

    Br. J. Exp. Pathol.

    (1988)
  • T. Solomon et al.

    Neurovirulence and host factors in flavivirus encephalitis-evidence from clinical epidemiology

    Arch. Virol.

    (2004)
  • A. Mathur et al.

    Host defense mechanisms against Japanese encephalitis virus infection in mice

    J. Gen. Virol.

    (1983)
  • K. Murali-Krishna et al.

    Protection of adult but not newborn mice against intracerebral challenge with Japanese encephalitis virus by adoptively transferred virus specific T lymphocytes: requirement of L3T4+ cells

    J. Gen. Virol.

    (1996)
  • C. Ramakrishna et al.

    T helper responses to Japanese encephalitis virus infection are dependent on the route of inoculation and the strain of mouse used

    J. Gen. Virol.

    (2003)
  • P. Kumar et al.

    Conserved amino acids 193-324 of non-structural protein 3 are a dominant source of peptide determinants for CD4+ and CD8+ T cells in a healthy Japanese encephalitis virus-endemic cohort

    J. Gen. Virol.

    (2004)
  • E. Konishi et al.

    The anamnestic neutralizing antibody response is critical for protection of mice from challenge following vaccination with a plasmid encoding the Japanese encephalitis virus premembrane and envelope genes

    J. Virol.

    (1999)
  • S. Srivastava et al.

    Degradation of Japanese encephalitis virus by neutrophils

    Int. J. Exp. Pathol.

    (1999)
  • T. Lee et al.

    Immune response in mice infected with the attenuated Japanese encephalitis vaccine strain SA14-14-2

    Acta Virol.

    (1995)
  • Y.L. Lin et al.

    DNA immunization with Japanese encephalitis virus non-structural protein NS-1 elicits protective immunity in mice

    J. Virol.

    (1998)
  • M. Hassert et al.

    Identification of protective CD8 T cell responses in a mouse model of Zika virus infection

    Front. Immunol.

    (2018)
  • K. Miura et al.

    A single gene controls resistance to Japanese encephalitis in mice

    Arch. Virol.

    (1990)
  • M. Hassert et al.

    CD4+ T cells mediate protection against Zika associated severe disease in a mouse model of infection

    PLoS Pathog.

    (2018)
  • C.H. Pan et al.

    Protective mechanisms induced by a Japanese encephalitis virus vaccine: requirement for antibody but not CD8+ cytotoxic T cell responses

    J. Virol.

    (2001)
  • D.S. Burke et al.

    Levels of interferon in the plasma and cerebrospinal fluid of patients with acute Japanese encephalitis

    J. Infect. Dis.

    (1987)
  • V. Ravi et al.

    Correlation of tumor necrosis factor levels in the serum and cerebrospinal fluid with clinical outcome in Japanese encephalitis patients

    J. Med. Virol.

    (1997)
  • A. Singh et al.

    Secretion of the chemokine IL-8 during Japanese encephalitis virus infection

    J. Med. Microbiol.

    (2000)
  • P.M. Winter et al.

    Proinflammatory cytokines and chemokines in humans with Japanese encephalitis

    J. Infect. Dis.

    (2004)
  • A. Ghoshal et al.

    Proinflammatory mediators released by activated microglia induces neuronal death in Japanese encephalitis

    Glia

    (2007)
  • U.C. Chaturvedi et al.

    Variable effects on peripheral blood leukocytes during JE virus infection of man

    Clin. Exp. Immunol.

    (1979)
  • R.T. Johnson et al.

    Japanese encephalitis: immunocytochemical studies of viral antigen and inflammatory cells in fatal cases

    Ann. Neurol.

    (1985)
  • V.J. Quagliarello et al.

    Recombinant human interleukin-1 induces meningitis and blood-brain barrier injury in the rat

    J. Clin. Investig.

    (1991)
  • M. Miyake

    The pathology of Japanese encephalitis

    Bull. World Health Organ.

    (1964)
  • E.G. Westaway et al.

    Flaviviridae

    Intervirology

    (1985)
  • S. Mukhopadhyay et al.

    A structural perspective of the flavivirus life cycle

    Nat. Rev. Microbiol.

    (2005)
  • T. Solomon et al.

    Japanese encephalitis

    J. Neurol. Neurosurg. Psychiatry

    (2000)
  • S. Vrati et al.

    Complete nucleotide sequence of an Indian strain of Japanese encephalitis virus: sequence comparison with other strains and phylogenetic analysis

    Am. J. Trop. Med. Hyg.

    (1999)
  • T.J. Chambers et al.

    Flavivirus genome organization, expression and replication

    Annu. Rev. Microbiol.

    (1990)
  • K. Stadler et al.

    Proteolytic activation of tick-borne encephalitis virus by furin

    J. Virol.

    (1997)
  • T. Solomon

    Viral encephalitis in Southeast Asia

    Neurol. Infect. Epidemiol.

    (1997)
  • Cited by (19)

    • Feasibility of chitosan-based nanoparticles approach for intranasal immunisation of live attenuated Japanese encephalitis vaccine

      2021, International Journal of Biological Macromolecules
      Citation Excerpt :

      However, the limitations of parenteral immunisation, including high costs and poor compliance, have also been reported. Consequently, intranasal (IN) immunisation of live attenuated influenza vaccine (Flu Mist®, Med Immune Vaccines Inc., US), a non-invasive mucosal route, is being explored and developed as an alternative route to overcome the disadvantages of parenteral immunisation [5]. Moreover, it is currently available on the market [6,7].

    View all citing articles on Scopus
    View full text