HEV Mosaic

Hepatitis E Virus Mosaic Recombinant
Shipped with Ice Packs
In Stock

Description

The e.coli derived HEV Mosaic protein contains 12 HEV immunodominant regions from ORF2 and ORF3 having a molecular mass of 38.5 kDa.

Product Specs

Introduction
Hepatitis E virus (HEV) is the primary cause of enterically transmitted non-A, non-B hepatitis globally. This spherical, non-enveloped, single-stranded RNA virus, measuring roughly 32 to 34 nm in diameter, is classified within the HEV-like viruses genus (unassigned). HEV possesses a single-stranded polyadenylated RNA genome of approximately 8 kb and is thought to be a calici-like virus based on its physicochemical characteristics.
Description
The HEV Mosaic protein, derived from e.coli, comprises 12 immunodominant HEV regions from ORF2 and ORF3, resulting in a molecular mass of 38.5 kDa.
Purity
The purity of HEV Mosaic protein exceeds 95%, as assessed by 10% PAGE (coomassie staining).
Formulation
The formulation consists of 25mM Tris pH 8.0, 1mM EDTA, 0.5M urea, and 50% glycerol.
Stability
While HEV Mosaic protein remains stable at 4°C for up to one week, storage below -18°C is recommended. Avoid repeated freeze-thaw cycles.
Purification Method
GS-4B Sepharose-Affinity Purification.
Specificity
Immunoreactive with sera HEV-infected individuals.

Q&A

What are the key structural components of HEV, and how do they relate to the mosaic approach in research?

The Hepatitis E Virus contains several important structural components organized in a mosaic-like arrangement. The virus has multiple open reading frames (ORFs) encoding various functional proteins. ORF1 encodes non-structural proteins including methyltransferase (MT), papain-like cysteine protease (PCP), helicase (Hel), and RNA-dependent RNA polymerase (RdRp) . When studying these components using a mosaic approach, researchers analyze the distinct functional domains and their interactions as interconnected elements of viral replication machinery.

Methodologically, this requires:

  • Domain mapping through sequence analysis

  • Functional characterization of individual domains

  • Integration of domain-specific data to understand the complete viral replication cycle

  • Study of interdomain interactions that may reveal cooperative functions

How does the structural mosaic of HEV domains facilitate viral evasion of host immune responses?

HEV employs multiple structural domains working in concert to evade host immunity. The methyltransferase domain prevents interferon regulatory factor 3 and the p65 subunit of NF-κB from phosphorylation and activation in a dose-dependent manner . Additionally, HEV MT strongly inhibits pattern recognition receptors including melanoma differentiation-associated protein 5 (MDA5) and RIG-I, which normally sense cytoplasmic double-stranded RNA and activate type 1 interferon responses .

To study these evasion mechanisms, researchers should:

  • Design domain-specific knockout experiments

  • Measure changes in immune signaling pathways using reporter assays

  • Quantify protein-protein interactions between viral and host factors

  • Examine temporal dynamics of immune evasion during the viral life cycle

What methodological approaches are most effective for constructing 3D models of HEV helicase?

Constructing reliable 3D models of HEV helicase involves several methodological considerations:

  • Template selection: The tomato mosaic virus (ToMV) helicase has been identified as the best template, sharing 33% structural identity with HEV helicase .

  • Model construction workflow:

    • Sequence alignment and template identification using functionally conserved domain search methods

    • Homology modeling using tools like Swiss-modeler

    • Validation through Ramachandran plot analysis (target: >85% residues in favored regions)

    • Further validation using Prosaweb server (optimal Z-score: approximately -6.4)

  • Molecular dynamics simulation:

    • Use GROMAC software with explicit solvent parameters

    • Assess stability through Root Mean Square Deviation (RMSD) analysis (stable value around 0.4 nm)

    • Evaluate residue mobility using Root Mean Square Fluctuation (RMSF)

This integrated approach produces models that accurately represent the structural and functional properties of HEV helicase for subsequent research applications.

How do researchers validate the accuracy of computational models of HEV proteins?

Validation of HEV protein models requires multiple complementary approaches:

  • Structural validation metrics:

    • Ramachandran plot analysis showing distribution of phi-psi angles (85.8% in favored regions for validated HEV helicase models)

    • Analysis of residues in additionally allowed regions (11.2%), generously allowed regions (0.5%), and disallowed regions (2.5%)

    • Z-score evaluation (averaged -6.4 for validated HEV helicase model)

  • Dynamic stability assessment:

    • RMSD analysis to track structural stability over simulation time (stabilization at ~10 ns)

    • RMSF analysis to identify regions of high mobility

    • Assessment of hydrogen bonding networks and salt bridges

  • Functional validation:

    • Docking studies with known ligands

    • Correlation with experimental mutagenesis data

    • Comparison with homologous proteins of known structure

The integration of these approaches ensures that computational models accurately represent both structural features and functional properties of HEV proteins.

What are the critical methodological considerations when designing mutational studies of HEV helicase?

When designing mutational studies of HEV helicase, researchers should consider:

  • Mutation strategy selection:

    • Saturation mutagenesis for comprehensive domain analysis

    • Site-directed mutagenesis for targeting specific functional motifs

    • Construction of chimeric proteins to isolate domain functions

  • Experimental readout systems:

    • GFP reporter replicons for real-time monitoring of replication effects

    • In vitro enzymatic assays for direct measurement of helicase activity

    • Protein expression systems for structure-function studies

  • Critical control design:

    • Include wild-type controls (e.g., pSK-GFP-WT)

    • Engineer known defective mutants as negative controls (e.g., pSK-GFP-GAD for RdRp mutants)

    • Design mutations that maintain protein structure while altering specific functions

  • Analysis of discrepancies between nucleotide and amino acid conservation:

    • Studies show that amino acid conservation in Walker motifs is critical for viability

    • Nucleotide sequences show more flexibility without affecting function

    • Design experiments that can distinguish these effects

How do researchers interpret seemingly contradictory results in HEV mutation studies?

When facing contradictory results in HEV mutation studies, researchers should implement the following analytical framework:

What is the optimal workflow for virtual screening of potential HEV helicase inhibitors?

The optimal workflow for virtual screening of HEV helicase inhibitors involves:

  • Target preparation:

    • Generate validated 3D model of HEV helicase (using homology modeling with ToMV helicase)

    • Identify binding pockets, with particular focus on Walker A and B motifs

    • Optimize model through molecular dynamics simulation

  • Compound library preparation:

    • Select diverse chemical databases (e.g., Maybridge)

    • Filter compounds for drug-like properties

    • Generate multiple conformers for flexible docking

  • Docking methodology:

    • Employ multiple docking programs for cross-validation (e.g., AutoDock Vina and GOLD 5.0)

    • Set appropriate parameters:

      • van der Waals annealing parameter: 5.0

      • Hydrogen bonding annealing parameter: 2.5

      • Genetic algorithm parameters: population size 100, selection pressure 1.2, operations 100,000

  • Post-docking analysis:

    • Score and rank compounds based on binding energy and interactions

    • Prioritize compounds interacting with critical residues (e.g., Walker A residues Gly975, Gly978, Ser979, Gly980)

    • Evaluate drug-likeness properties

  • Validation and refinement:

    • Re-dock top hits with more stringent parameters

    • Perform molecular dynamics simulations on protein-ligand complexes

    • Select candidates for experimental validation

This comprehensive workflow has identified promising inhibitors including JFD02650, RDR03130, and HTS11136, which interact with critical Walker A residues .

How can researchers integrate structural insights with functional data to identify novel HEV inhibitor binding sites?

Integration of structural and functional data for identification of novel HEV inhibitor binding sites requires:

  • Structural domain mapping with functional correlation:

    • Identify conserved motifs (Walker A, Walker B, motifs Ia and III)

    • Map mutagenesis data onto structural models

    • Correlate binding sites with enzymatic activities (ATPase, unwinding)

  • Hotspot identification techniques:

    • Computational solvent mapping

    • Fragment-based screening simulations

    • Analysis of residue conservation across HEV genotypes

    • Identification of allosteric sites through normal mode analysis

  • Integration of experimental data:

    • Mutations L1110F and V1120I downstream of Walker B decrease ATPase activity

    • Deletions in motifs Ia and III impair ATPase and unwinding activities

    • Mutations in Walker A and B motifs abolish replication

  • Innovative binding site characterization:

    • The W437-W476/rs5 domain in HEV protease interacts with proline-rich regions

    • Metal-binding motifs contribute to structural integrity

    • Targeting protein-protein interaction interfaces between viral domains

This integrated approach identifies functional hotspots that may not be obvious from structure alone, providing novel targets for antiviral development.

What are the strengths and limitations of current replicon systems for studying HEV mutations?

HEV replicon systems offer crucial tools for studying viral mutations, with specific strengths and limitations:

AspectStrengthsLimitations
DesignGFP reporters allow real-time monitoringReporter genes may affect replication kinetics
Mutation analysisPermits systematic study of domain functionsMay not reflect effects in complete viral lifecycle
Host systemsEnables study in relevant cell linesLimited to permissive cell types
ControlsIncludes validated controls like RdRp mutantsCell culture adaptation may introduce artifacts
Application rangeEffective for studying replication machineryLimited for studying assembly and release
ThroughputAllows screening of multiple mutationsResource-intensive for comprehensive studies

For comprehensive analysis, researchers should:

  • Confirm replicon findings using multiple cell lines

  • Validate key findings in infectious virus systems when possible

  • Consider the impact of reporter gene insertion on viral RNA structure

  • Account for cell culture adaptations that may not reflect in vivo behavior

How can researchers design experiments to distinguish between effects on RNA structure versus protein function in HEV mutational studies?

Designing experiments to differentiate between RNA structural effects and protein functional effects requires:

  • Strategic mutation design:

    • Silent mutations: Alter nucleotide sequence while preserving amino acid sequence

    • Synonymous codon substitutions: Change codons without changing amino acids

    • Compensatory mutations: Restore RNA structures while maintaining amino acid changes

  • Comparative mutation analysis:

    • Design parallel mutations that affect either nucleotide or amino acid sequence exclusively

    • Compare saturation mutagenesis (affecting nucleotides) with site-directed mutagenesis (targeting amino acids)

  • RNA structure analysis:

    • SHAPE (Selective 2'-hydroxyl acylation analyzed by primer extension)

    • Dimethyl sulfate (DMS) probing

    • RNA structure prediction algorithms

  • Protein function assays:

    • In vitro ATPase activity assays

    • RNA unwinding assays

    • Protein-protein interaction studies

  • Integrated analysis:

    • Correlate RNA structural changes with replication efficiency

    • Map protein functional changes to specific biochemical activities

    • Use molecular dynamics simulations to model effects of mutations

Research shows that while saturation mutations across the HEV helicase domain did not significantly affect replication, specific amino acid changes in Walker motifs abolished replication completely, indicating the primacy of protein function over RNA structure in this context .

How do the structural features of HEV helicase compare with other viral helicases, and what are the implications for broad-spectrum antiviral design?

Comparative analysis of HEV helicase with other viral helicases reveals:

  • Structural conservation patterns:

    • HEV helicase shares 33% structural identity with tomato mosaic virus (ToMV) helicase

    • Conservation of Walker A and B motifs across positive-sense RNA viruses

    • Shared "papain-like β-barrel fold" in protease domains

  • Functional domain organization:

    • Core helicase domain with conserved motifs for ATP binding and hydrolysis

    • RNA-binding regions with variable sequence specificity

    • Integration with other viral proteins through specific interaction domains

  • Implications for broad-spectrum antiviral design:

    • Target highly conserved residues in Walker motifs (Gly975, Gly978, Ser979, Gly980)

    • Design inhibitors that interact with structural elements conserved across viral families

    • Exploit unique features of viral helicases distinct from human counterparts

The 3D model of HEV helicase created using ToMV as a template provides a foundation for comparing viral helicases and identifying both conserved and unique features that can be exploited for antiviral development.

What methodological approaches are most effective for studying the interactions between HEV helicase and other viral/host proteins?

Effective methodological approaches for studying HEV helicase interactions include:

  • Protein-protein interaction detection methods:

    • Co-immunoprecipitation followed by mass spectrometry

    • Yeast two-hybrid screening

    • Proximity-based labeling (BioID, APEX)

    • Förster resonance energy transfer (FRET)

  • Interaction domain mapping:

    • Deletion mutant analysis

    • Domain swapping experiments

    • Site-directed mutagenesis of interface residues

    • Peptide competition assays

  • Structural characterization:

    • X-ray crystallography of protein complexes

    • Cryo-electron microscopy

    • Nuclear magnetic resonance (NMR) for dynamic interactions

    • Computational docking validated by experimental data

  • Functional validation:

    • Mutations in interaction interfaces

    • Competitive inhibition using peptides or small molecules

    • Correlation of interaction disruption with functional effects

  • Host-protein interactions:

    • Study interactions with MDA5 and RIG-I pathways

    • Analyze effects on interferon regulatory factor 3 and NF-κB signaling

    • Investigate interactions with calcium/calmodulin pathways suggested by Ca²⁺/CaM-binding motifs

How can researchers leverage the identification of metal-binding motifs in HEV for novel therapeutic approaches?

The discovery of metal-binding motifs in HEV proteins opens several research avenues for therapeutic development:

  • Characterizing metal-binding domains:

    • The HEV PCP domain contains Zn²⁺-binding motifs essential for structural integrity

    • Ca²⁺/CaM-binding motifs with intramolecular disulfide bridges orient EF-hands toward Ca²⁺ binding

    • These metal-binding regions are crucial for polyprotein processing and RNA replication

  • Therapeutic targeting strategies:

    • Metal chelators that selectively disrupt viral protein function

    • Small molecules that alter metal coordination geometry

    • Compounds that compete for metal-binding sites

    • Allosteric modulators that affect metal-binding domain dynamics

  • Methodological approaches:

    • Metal-binding site prediction algorithms

    • Isothermal titration calorimetry to measure binding affinities

    • X-ray absorption spectroscopy to characterize metal coordination

    • Structure-based design of metal-binding site inhibitors

  • Experimental validation:

    • Site-directed mutagenesis of metal-coordinating residues

    • Functional assays with metal chelators and competitors

    • Structural studies of metal-bound versus apo protein forms

This research direction exploits the dependence of HEV proteins on metal cofactors for structural integrity and catalytic function, potentially leading to novel classes of antivirals with distinct mechanisms of action.

What are the methodological challenges in studying the dynamic conformational changes of HEV helicase during RNA unwinding?

Studying dynamic conformational changes in HEV helicase during RNA unwinding presents several methodological challenges:

  • Technical challenges in capturing transient states:

    • RNA unwinding occurs through rapid conformational changes

    • Intermediate states are often unstable and difficult to trap

    • ATP hydrolysis and RNA binding/release are temporally coupled events

  • Advanced methodological approaches:

    • Single-molecule FRET to monitor real-time conformational changes

    • Time-resolved X-ray crystallography with temperature-jump initiation

    • Hydrogen-deuterium exchange mass spectrometry

    • Molecular dynamics simulations with enhanced sampling techniques

  • Experimental system design:

    • Construction of partial reaction systems to isolate specific steps

    • Design of RNA substrates with fluorescent or FRET pairs

    • Use of ATP analogs to trap specific conformational states

    • Development of HEV helicase variants with altered kinetics

  • Integration with structural data:

    • Combine static structural information from crystallography

    • Incorporate molecular dynamics simulations to model transitions

    • Validate computational models with experimental measurements

    • Build Markov state models of the complete unwinding process

Understanding these dynamic processes is essential for designing inhibitors that target specific conformational states or transitions rather than just the ground state structure of the enzyme.

How can the structural modeling and inhibitor identification approaches for HEV helicase be optimized for clinical translation?

Optimizing HEV helicase research for clinical translation requires:

  • Refinement of structural models for drug discovery:

    • Increase model accuracy through integration of experimental data

    • Validate functional importance of binding sites through mutagenesis

    • Characterize flexibility and conformational states relevant to drug binding

  • Inhibitor optimization workflow:

    • Screen virtual hits against multiple HEV genotypes

    • Optimize compounds for pharmacokinetic properties

    • Address toxicity concerns through medicinal chemistry optimization

    • Develop structure-activity relationships guided by binding mode analysis

  • Resistance barrier assessment:

    • Identify potential resistance mutations using structural analysis

    • Design inhibitors that target highly conserved residues (e.g., Walker A motifs)

    • Develop combination approaches targeting multiple viral functions

    • Model effects of known natural variations on inhibitor binding

  • Translational research considerations:

    • Develop cell-based assays that reflect in vivo conditions

    • Establish animal models for pharmacokinetic/pharmacodynamic studies

    • Address formulation and delivery challenges

    • Consider combination with existing therapies (pegIFN-α-2a, ribavirin)

This systematic approach builds on the basic research findings to develop clinically viable therapeutic candidates with optimal efficacy and resistance profiles.

What are the critical considerations when interpreting the clinical relevance of naturally occurring HEV helicase mutations?

When interpreting the clinical relevance of naturally occurring HEV helicase mutations, researchers should consider:

  • Correlation with disease severity:

    • L1110F and V1120I mutations (downstream of Walker B) are detected in fulminant liver failure patients

    • These mutations decrease ATPase activity and RNA replication in vitro

    • Suggests functional significance of helicase activity in pathogenesis

  • Analytical framework:

    • Distinguish between primary pathogenic mutations and compensatory changes

    • Assess effects on multiple viral functions (replication, immune evasion)

    • Consider host genetic background and comorbidities

    • Evaluate frequency across viral genotypes and geographic regions

  • Methodological validation:

    • Confirm mutations in multiple patient samples

    • Reconstruct mutations in laboratory strains

    • Measure effects on multiple aspects of viral fitness

    • Assess stability of mutations during transmission and chronic infection

  • Clinical implications:

    • Potential for personalized antiviral approaches

    • Prognostic value of specific mutations

    • Impact on treatment response to standard therapies

    • Contribution to epidemic potential and transmission dynamics

Understanding the functional consequences of naturally occurring mutations provides insights into viral pathogenesis and can guide the development of diagnostic tools and therapeutic strategies.

Product Science Overview

Introduction

Hepatitis E virus (HEV) is a significant cause of acute hepatitis worldwide, particularly in developing countries. It is a single-stranded, positive-sense RNA virus that primarily spreads through the fecal-oral route, often via contaminated water. HEV infections can lead to severe liver disease, especially in pregnant women and immunocompromised individuals .

Hepatitis E Virus Structure and Genotypes

HEV belongs to the Hepeviridae family and has four primary genotypes that infect humans: HEV-1, HEV-2, HEV-3, and HEV-4. Genotypes 1 and 2 are typically found in developing countries and are associated with large outbreaks, while genotypes 3 and 4 are zoonotic and can be transmitted from animals to humans .

Recombinant HEV Vaccines

The development of recombinant HEV vaccines has been a significant advancement in preventing HEV infections. These vaccines are based on the HEV open reading frame 2 (ORF2) capsid protein, which is crucial for the virus’s ability to infect host cells. The ORF2 protein is often produced using recombinant DNA technology in various expression systems, such as Escherichia coli .

Mosaic Recombinant HEV Vaccines

Mosaic recombinant HEV vaccines are designed to enhance the immune response by incorporating multiple epitopes from different HEV genotypes. This approach aims to provide broader protection against various HEV strains. The mosaic design involves creating chimeric proteins that combine antigenic regions from different HEV genotypes, thereby eliciting a more robust and cross-protective immune response .

Preparation Methods

The preparation of mosaic recombinant HEV vaccines involves several steps:

  1. Gene Cloning: The genes encoding the desired antigenic regions from different HEV genotypes are cloned into suitable expression vectors.
  2. Protein Expression: The recombinant vectors are introduced into expression systems, such as E. coli, to produce the chimeric proteins.
  3. Protein Purification: The expressed proteins are purified using techniques like affinity chromatography to obtain high-purity antigens.
  4. Formulation: The purified proteins are formulated with adjuvants to enhance the immune response and stability of the vaccine .
Efficacy and Safety

Clinical trials have demonstrated the efficacy and safety of recombinant HEV vaccines. For instance, a study showed that a recombinant HEV vaccine based on the ORF2 protein had an efficacy of 86.8% over a 4.5-year period . The vaccine was well-tolerated, with no significant adverse effects reported .

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.