JEV

Japanese Encephalitis Virus ENV Recombinant
Shipped with Ice Packs
In Stock

Description

Genome and Replication

The JEV genome lacks a poly-A tail but includes a 5′ methylated cap. A single open reading frame (ORF) translates into a polyprotein cleaved by viral/host proteases into functional subunits . NS3 helicase and NS5 polymerase are critical for RNA replication .

Pathogenesis and Host Interactions

JEV primarily targets neurons and endothelial cells, causing neuroinflammation, vasculitis, and blood-brain barrier (BBB) disruption . Key mechanisms include:

  • BBB Breakdown: Occludin degradation in tight junctions facilitates viral entry into the CNS .

  • Immune Evasion: NS1 and NS2A proteins inhibit AMPK activation, promoting lipid synthesis for viral replication .

  • Envelope Protein Dynamics: The E protein’s EDIII domain binds host receptors (e.g., HSP70, GRP78), while prM cleavage in the Golgi exposes fusion peptides for mature virion release .

Promising Candidates and Mechanisms

CompoundTargetEC₅₀/CC₅₀Mechanism
Isobavachalcone (IBC)AMPK activationEC₅₀: ~23–25 µMRestores AMPK signaling, inhibits lipid synthesis
Corosolic acid (CA)AMPK activationEC₅₀: ~23–25 µMBlocks JEV NS1/NS2A-mediated AMPK suppression
FGIN-1-27UnknownEC₅₀: 3.21 µMInhibits viral replication phase
CilnidipineUnknownEC₅₀: 6.52 µMSelectively blocks JEV proliferation
CW-33 analoguesNS2B-NS3 proteaseIC₅₀: 3.21–6.52 µMBinds Arg76, Glu155 via H-bonds and hydrophobic interactions
  • NS3 Helicase Inhibitors: Two compounds (EC₅₀: ~24 µM) disrupt RNA unwinding without affecting ATPase activity, reducing viral titers in vitro .

  • QSAR-Optimized Analogues: CW-33 derivatives with meta-substituted phenyl groups show enhanced antiviral activity via molecular docking simulations .

Challenges and Future Directions

Despite progress, no JEV-specific antivirals are clinically approved. Key hurdles include:

  • Drug Delivery: Crossing the BBB remains problematic for CNS-targeted therapies .

  • Broad-Spectrum Potential: Compounds like IBC and CA inhibit multiple viruses (Zika, coronaviruses), suggesting utility beyond JE .

  • Resistance Risks: Targeting conserved regions (e.g., NS3 helicase) may reduce escape mutations .

Epidemiological Impact

  • Annual Cases: ~100,000 globally, with 25,000 deaths .

  • At-Risk Populations: Children in endemic regions (Asia), though adults lack immunity in non-endemic areas .

Product Specs

Introduction
Japanese encephalitis, formerly known as Japanese B encephalitis, is a viral infection from the Flaviviridae family. Closely related to the West Nile and St. Louis encephalitis viruses, it is characterized by a positive-sense, single-stranded RNA genome encased in a capsid protein shell. The virus's outer envelope, comprised of the envelope (E) protein, acts as a protective antigen and facilitates cellular entry. Beyond structural components, the genome encodes nonstructural proteins (NS1, NS2a, NS2b, NS3, N4a, NS4b, NS5), with NS1 also existing in a secreted form. Notably, NS3 functions as a putative helicase, while NS5 serves as the viral polymerase. Japanese encephalitis virus (JEV) is known to infect the endoplasmic reticulum (ER) lumen, where it rapidly accumulates viral proteins. Diagnosis typically involves detecting antibodies in serum and cerebrospinal fluid (CSF) using IgM capture ELISA.
Description
This recombinant protein, derived from E. coli, encompasses the full-length Japanese Encephalitis virus ENV antigen, exhibiting a molecular weight of 50 kDa. It is engineered with a 6-histidine tag. GenBank Accession Number: AHK05344.1
Purity
The Encephalitis protein demonstrates a purity greater than 90%, as determined by SDS-PAGE analysis with Coomassie blue staining.
Formulation
The protein is supplied in a buffer consisting of 20mM Tris-MES (pH 6.5), 8M urea, 200mM NaCl, and 0.05% Tween-20.
Stability
For optimal stability, the Encephalitis protein should be stored at temperatures below -18°C. While it can remain stable at 4°C for up to one week, it is advisable to minimize freeze-thaw cycles.
Source
Escherichia Coli.
Purification Method
Encephalitis protein was Purified by proprietary chromatographic technique.
Specificity

Immunoreactive with sera from JEV-infected individuals.

Q&A

What is JEV and what is its genomic structure?

JEV is a flavivirus in the family Flaviviridae that causes Japanese encephalitis, a serious neurological disease. The virus has a single-stranded, positive-sense RNA genome of approximately 11 kb that encodes a single polyprotein, which is subsequently cleaved into three structural proteins (capsid, pre-membrane, and envelope) and seven non-structural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5) . Each protein plays specific roles in viral replication, assembly, and immune evasion.

How are JEV genotypes classified and what is their geographical distribution?

JEV is classified into five genotypes (1-5) based on genetic diversity primarily in the envelope protein gene. Genotypes 1-3 are more prevalent across Asia, while genotype 4 has been recently detected in Australia in an unprecedented outbreak . Genotype 5 is the rarest. These genotypes show varying degrees of virulence and host interactions, with genotype 4 demonstrating lower virulence in experimental models compared to genotypes 2 and 3 .

What is the significance of NS3 helicase in JEV replication?

The C-terminus of non-structural protein 3 (NS3) encodes helicase functionality, which is essential for viral RNA replication. This domain unwinds double-stranded RNA intermediates during viral genome replication. Due to its critical role, NS3 helicase has been identified as a potential drug target . The protein contains an RNA unwinding channel that, when blocked by inhibitors, can prevent viral replication without affecting its ATPase activity, making it an attractive target for antiviral development.

How does JEV genotype 4 structurally differ from other genotypes?

JEV genotype 4, recently isolated in Australia (JEV NSW/22), shows structural differences primarily in its envelope protein. These differences affect the virus's interactions with host immune systems. Molecular studies indicate that compared to genotype 2 (JEV FU) and genotype 3 (JEV Nakayama), JEV genotype 4 displays increased sensitivity to type I interferon responses, which may partially explain its lower virulence in certain mouse models .

What animal models are most effective for studying JEV pathogenesis?

Several mouse models have demonstrated utility in JEV research:

Mouse ModelCharacteristicsResearch Applications
C57BL/6J (wild-type)Less susceptible to JEV infectionInitial screening, mild disease models
IRF7-deficientMore susceptible than wild-typeNeuroinvasion studies, higher viremia
IFNAR-deficientUniversally lethal by day 3Severe disease models, hemorrhagic manifestations

IRF7-deficient mice provide a particularly useful model for JEV NSW/22 (genotype 4), showing higher viremia levels compared to wild-type mice and allowing for lethal neuroinvasive infection studies . All JEV strains are universally lethal in IFNAR-deficient mice by day 3, with histological signs of brain hemorrhage.

How can human cortical brain organoids advance JEV neurotropism research?

Human cortical brain organoids (hBOs) provide a valuable in vitro system for studying JEV neurotropism and pathogenesis. Research demonstrates that all JEV isolates can establish robust cytopathic infection in hBOs, although genotype 4 (JEV NSW/22) shows lower infection rates . This model allows researchers to study:

  • Virus-neural cell interactions in a human-specific context

  • Mechanisms of neuronal damage and cell death

  • Host immune responses in neural tissues

  • Comparative virulence of different JEV genotypes without the species barrier limitations of animal models

What screening methodologies are most effective for identifying anti-JEV compounds?

High-throughput molecular docking has proven effective in identifying potential JEV inhibitors, particularly those targeting the NS3 helicase. In one study, researchers screened a commercial library containing 250,000 compounds against the JEV NS3 helicase structure, identifying 41 promising candidates that were subsequently tested for their ability to inhibit NS3 activity . Two compounds were found to strongly inhibit unwinding activity without affecting ATPase function, demonstrating EC50 values of 25.67 and 23.50 μM, respectively.

How can structure-based drug design be applied to develop JEV-specific inhibitors?

Structure-based drug design for JEV inhibitors involves:

  • Identifying critical binding pockets in viral proteins (e.g., NS3 helicase RNA unwinding channel)

  • Using computational simulations to identify molecules that can effectively bind these targets

  • Testing compounds' inhibitory effects on specific viral functions (e.g., unwinding vs. ATPase activity)

  • Confirming antiviral activity in cell culture through assays like Western blots, immunofluorescence, and plaque reduction

  • Identifying specific atoms participating in intramolecular interactions to guide compound optimization

This approach has successfully identified compounds that bind and block the NS3 RNA unwinding channel, consistent with their ability to inhibit unwinding activity without affecting ATPase function.

How might existing vaccination approaches against related flaviviruses be leveraged against JEV?

Recent research suggests potential cross-protection between related flaviviruses. Investigations are underway to determine if West Nile Virus vaccination could provide protection against JEV, as good cross-immunity exists between these viruses . This approach could provide quick and readily available resources to swine producers in emergency situations, such as outbreaks in previously non-endemic regions like the United States.

How do different mosquito species vary in their capacity to transmit JEV?

Mosquito vector competence for JEV transmission depends on multiple factors including:

  • Ability to support viral replication

  • Presence of suitable receptors for viral entry

  • Overcoming midgut and salivary gland barriers

  • Environmental conditions affecting mosquito-virus interactions

Research is currently investigating the capability of different mosquito species to carry, acquire, and transmit various JEV genotypes . This information is crucial for predicting transmission dynamics in different geographical regions and developing targeted vector control strategies.

What methodologies are most effective for assessing vector competence experimentally?

Experimental assessment of vector competence typically involves:

  • Exposing mosquitoes to JEV through infected blood meals at controlled viral titers

  • Incubating mosquitoes under standardized temperature and humidity conditions

  • Sampling mosquitoes at various time points to test for:

    • Virus presence in midgut (infection)

    • Virus dissemination to secondary tissues

    • Virus presence in saliva (transmission potential)

  • Quantifying viral load using plaque assays or RT-PCR

  • Comparing transmission rates across mosquito species and viral genotypes

What factors influence formaldehyde inactivation efficiency for JEV vaccines?

Formaldehyde inactivation, a common method for inactivated JEV vaccines, is affected by several key variables:

  • Temperature

  • pH

  • Formaldehyde concentration

  • Presence of stabilizers (glycerol, sorbitol, lysine, glycine, and PEG)

These factors significantly impact both viral inactivation kinetics and antigen recovery rates. Due to the protein cross-linking mechanism by which formaldehyde inactivation occurs, antigen recovery rates can vary considerably, affecting vaccine potency and effectiveness.

How can Design of Experiments (DoE) methodology optimize JEV inactivation processes?

Design of Experiments approaches for optimizing JEV inactivation include:

  • Two-level factorial designs to screen multiple variables simultaneously

  • Center points to assess curvature in the response surface

  • Measured outcomes including:

    • Antigen recovery by ELISA

    • Virion size measurements

    • Complete inactivation verification by plaque assay

This methodology allows researchers to determine which factors are most significant and which contribute to interaction effects for optimal JEV inactivation in vaccine production.

What surveillance strategies are most effective for early detection of JEV emergence?

Effective JEV surveillance combines multiple approaches:

  • Sentinel animal monitoring, particularly in pig populations

  • Vector surveillance with molecular testing

  • Human case detection through clinical surveillance

  • Serological surveys in high-risk populations

  • Genomic surveillance to track viral evolution

For regions at risk of JEV introduction, surveillance should focus on high-risk entry points such as international airports and seaports, combined with monitoring of suitable vectors and amplifying hosts in surrounding areas.

What are the gold standard methods for laboratory confirmation of JEV?

Laboratory diagnosis of JEV infection relies on several complementary methods:

  • Serological testing (IgM capture ELISA in cerebrospinal fluid)

  • Molecular detection via RT-PCR

  • Virus isolation in cell culture with immunofluorescence confirmation

  • Lumbar puncture (spinal tap) for cerebrospinal fluid analysis

When evaluating patients with suspected JEV infection, clinicians should consider recent travel history to areas with JEV cases and the presence of characteristic neurological symptoms.

How can researchers address the challenge of cross-reactivity in JEV serological diagnosis?

Cross-reactivity among flaviviruses presents significant challenges for serological diagnosis. Researchers can overcome this through:

  • Development of antibody tests targeting virus-specific epitopes

  • Implementation of virus neutralization tests as confirmatory assays

  • Use of multiplex assays that can differentiate between closely related flaviviruses

  • Inclusion of appropriate controls to detect potential cross-reactive antibodies

  • Integration of clinical and epidemiological data with laboratory results for accurate interpretation

How is whole genome sequencing advancing our understanding of JEV evolution?

Whole genome sequencing enables:

  • Precise genotyping of JEV isolates

  • Identification of novel mutations potentially associated with altered virulence

  • Tracking of geographical spread and evolutionary relationships

  • Detection of recombination events between genotypes

  • Assessment of selection pressures on viral proteins

This approach has been instrumental in characterizing the JEV genotype 4 strain involved in the recent Australian outbreak, providing insights into its evolutionary history and potential adaptation to new ecological niches .

What new approaches show promise for developing universal JEV antivirals?

Promising approaches for universal JEV antivirals include:

  • Targeting highly conserved viral proteins (like NS3 helicase) that are essential across all genotypes

  • Developing host-directed therapies that target cellular factors required for viral replication

  • Combination therapies that target multiple viral lifecycle stages

  • Repurposing existing broad-spectrum antivirals with activity against related flaviviruses

  • Exploring RNA interference and CRISPR-based strategies to inhibit viral replication

Product Science Overview

Introduction

Japanese Encephalitis Virus (JEV) is a mosquito-borne flavivirus that is a significant cause of viral encephalitis in Asia. It belongs to the same genus as dengue, Zika, yellow fever, and West Nile viruses . The first documented case of Japanese encephalitis was in 1871 in Japan . The virus is primarily transmitted by Culex mosquitoes, and it predominantly affects rural and agricultural areas where these mosquitoes breed.

Structure and Function

The JEV virion comprises three structural proteins: the envelope glycoprotein (E), the membrane protein (M), and the capsid protein © . The envelope glycoprotein (E) is the major determinant of the virus’s pathogenicity and host immune responses . It plays a crucial role in the virus’s ability to infect host cells and is the primary target for neutralizing antibodies.

Recombinant JEV E Protein

Recombinant technology has enabled the production of the JEV E protein in various expression systems. One notable approach involves the use of a Drosophila expression system. In this method, the coding sequence for the signal sequence of the premembrane and E protein is cloned into a Drosophila expression vector . The resulting recombinant E protein is immunoreactive and can be used for vaccine development and diagnostic purposes .

Vaccine Development

The development of vaccines against JEV has been a critical public health priority. The earliest vaccines were mouse brain-derived formalin-inactivated viruses . However, these vaccines had limitations, including production challenges and potential safety concerns. Recombinant vaccines, such as those based on the JEV E protein, offer a safer and more efficient alternative . These vaccines can elicit strong immune responses and provide long-term protection against the virus .

Epidemiology and Impact

JEV is the leading cause of viral encephalitis, with approximately 50,000 cases reported annually worldwide . The disease has a high mortality rate, with around 15,000 deaths each year, and up to 50% of survivors suffer from severe neuropsychiatric sequelae . Most cases occur in southern and eastern Asia, but the geographical range of JEV is expanding, with outbreaks reported in regions such as the Saipan Islands, Torres Straits Islands, and mainland Australia .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.