HEV ORF2 (633-659 a.a.)

Hepatitis E Virus ORF2 (633-659 a.a.) Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Sequence Context

The HEV ORF2 protein is predominantly intrinsically disordered, with significant regions lacking fixed 3D structures . Homology modeling reveals a mix of alpha-helices, beta-strands, and random coils, with beta-strands constituting ~36–39% of the protein . The 633–659 a.a. region is positioned near the C-terminus, a segment critical for capsid assembly and viral particle formation .

FeatureDetailsSource
Sequence Length660 a.a. (full ORF2); 633–659 a.a. is the terminal ~27 a.a. segment.
Secondary StructureBeta-strands dominate in C-terminal regions (36–39% in modeled structures).
Disorder PredictionModerately disordered (19–27% disordered residues in some genotypes).

Capsid Assembly and Stability

  • The C-terminal region (post-600 a.a.) may stabilize interactions between ORF2 subunits, critical for virion formation. Mutations in glycosylation sites (e.g., Asn-310, Asn-562) disrupt virion assembly , implying similar constraints for terminal regions.

  • Beta-strands in this region could mediate protein-protein interactions, analogous to structural roles in other viral capsid proteins.

Immune Evasion and Host Interactions

  • The N-terminal arginine-rich motif (1–111 a.a.) inhibits host interferon responses , but terminal regions may modulate immune recognition.

  • The 633–659 a.a. segment could influence antigen presentation or antibody binding, though no direct epitopes have been mapped here.

Immunological Significance

The ORF2 protein is the primary target of neutralizing antibodies, with the E2s domain (459–606 a.a.) being highly immunogenic . While 633–659 a.a. is not explicitly cited as an epitope, its structural role may indirectly affect antibody recognition:

Antigenic RegionA.a. RangeFunctionSource
E2s Domain459–606Major target for neutralizing antibodies.
C-Terminal Segment633–659Hypothetical role in capsid stability.

Research Gaps and Future Directions

  • Structural Characterization: Cryo-EM or crystallography studies are needed to resolve the 633–659 a.a. region’s interactions.

  • Functional Mutagenesis: Site-directed mutagenesis could test the role of residues in this segment during virion assembly or immune evasion.

  • Host Interactions: Screening for binding partners (e.g., host receptors) may reveal novel mechanisms.

Product Specs

Introduction
Hepatitis E virus (HEV) is a major cause of enterically transmitted non-A, non-B hepatitis globally. This spherical, non-enveloped, single-stranded RNA virus, measuring approximately 32-34 nm in diameter, is classified within the HEV-like viruses genus (unassigned genus). HEV possesses a single-stranded polyadenylated RNA genome of about 8 kb and is thought to be a calici-like virus based on its physicochemical properties.
Description
This E. coli-derived HEV protein represents genotype-2 and includes a beta-galactosidase fusion at its N-terminus. It encompasses the HEV immunodominant region within ORF2, spanning amino acids 633-659.
Purity
HEV ORF2 Protein exhibits a purity exceeding 95%, as determined by 10% PAGE (Coomassie staining).
Formulation
The protein is supplied in a buffer containing 20mM Tris-HCl (pH 8), 8M urea, and 10mM B-ME.
Stability
For optimal stability, HEV ORF2 protein should be stored below -18°C. While it remains stable at 4°C for up to one week, repeated freeze-thaw cycles should be avoided.
Applications
HEV ORF2 antigen is suitable for use in ELISA and Western blot applications. It serves as an excellent antigen for HEV detection, demonstrating minimal specificity issues.
Purification Method
HEV ORF2 protein was purified by proprietary chromatographic technique.
Specificity
Immunoreactive with sera HEV-infected individuals.

Q&A

What is HEV ORF2 (633-659 a.a.) and what makes this region significant?

HEV ORF2 (633-659 a.a.) refers to a specific immunodominant region within the Hepatitis E virus capsid protein. This region is particularly significant because it is highly immunoreactive with sera from HEV-infected individuals, making it valuable for diagnostic applications . The E.coli-derived HEV protein containing this region is typically fused with beta-galactosidase at the N-Terminus, which helps with protein expression and purification while maintaining immunogenicity .

Methodologically, researchers working with this region should note that it represents a conserved epitope across different HEV genotypes, explaining its effectiveness in broad-spectrum HEV detection assays. Experimental approaches utilizing this region often involve recombinant protein expression in bacterial systems, followed by chromatographic purification to achieve >95% purity .

How does the ORF2 (633-659 a.a.) region compare structurally across different HEV genotypes?

Comparative sequence analysis reveals that the ORF2 capsid protein, including the 633-659 a.a. region, is one of the more conserved regions across HEV genotypes. Phylogenetic analyses show that in the more conserved regions of ORF2 (residues 105 to 458), the average pairwise distance between representative sequences falls below 0.5, indicating significant conservation despite genetic variability elsewhere in the genome .

To properly investigate cross-genotype conservation experimentally, researchers should perform:

  • Multiple sequence alignment of the 633-659 region across genotypes 1-4 and zoonotic strains

  • Epitope mapping using truncated recombinants to identify the minimal immunogenic sequence

  • Cross-reactivity testing of antibodies raised against this region with proteins from different genotypes

The methodological implication is that antibodies targeting this region may provide broader detection capability than those targeting more variable regions of the virus.

What are the optimal storage and handling conditions for recombinant HEV ORF2 (633-659 a.a.) protein?

Recombinant HEV ORF2 (633-659 a.a.) protein requires specific storage and handling protocols to maintain stability and immunoreactivity. The protein is typically formulated in 20mM Tris-HCl pH-8, 8M urea, and 10mM B-ME . While it remains stable at 4°C for approximately one week, long-term storage should be below -18°C .

Methodology for proper handling includes:

  • Avoiding repeated freeze-thaw cycles, which can cause protein degradation and loss of antigenic properties

  • Working with aliquots rather than the entire stock to prevent contamination and degradation

  • Performing quality control by SDS-PAGE (10% gels with Coomassie staining) to verify >95% purity before experimental use

  • Confirming immunoreactivity periodically through ELISA with confirmed positive sera

Researchers should note that preparation methods significantly impact protein performance; purification via proprietary chromatographic techniques has proven effective in maintaining the conformational epitopes necessary for immunoassay applications .

How do post-translational modifications affect the immunogenicity of the ORF2 (633-659 a.a.) region?

Post-translational modifications, particularly N-glycosylation, significantly influence ORF2 protein biology, though their specific impact on the 633-659 a.a. region requires careful interpretation. The complete ORF2 protein contains three highly conserved potential N-glycosylation sites (N1, N2, and N3), with experimental evidence confirming glycosylation at N1 and N3 but not N2 .

Research demonstrates that while N-glycosylation does not play a role in replication and assembly of infectious HEV particles, it substantially affects protein stability and antigenicity . The glycosylated forms of ORF2 (ORF2g/c) are remarkably stable proteins and are primary targets of patient antibodies .

Methodologically, researchers investigating this phenomenon should:

  • Generate glycosylation-site mutants (N→Q substitutions) of the region to assess the impact on antibody recognition

  • Compare recombinant proteins expressed in bacterial systems (non-glycosylated) versus eukaryotic systems (glycosylated)

  • Perform deglycosylation experiments using PNGase F or Endo H to determine if glycan shields mask important epitopes

These approaches will help distinguish between the intrinsic immunogenicity of the peptide sequence versus the contribution of glycan modifications.

What is the role of ORF2 (633-659 a.a.) in modulating host immune responses during HEV infection?

Recent research has uncovered a critical immunomodulatory role for the ORF2 protein, with potential implications for the 633-659 a.a. region. The ORF2 protein has been shown to counteract cell-intrinsic antiviral responses through multiple mechanisms . It interferes with inflammatory signaling pathways and antiviral signaling downstream of pattern recognition receptors, partially through direct interaction with TANK binding kinase 1 .

Additionally, the secreted form of ORF2 (ORF2s) inhibits antibody-mediated neutralization of HEV , suggesting an immune evasion function. This interference with neutralizing antibodies may create a favorable environment for viral persistence, particularly in chronic infections .

To experimentally investigate this region's immunomodulatory properties, researchers should:

  • Develop truncated ORF2 constructs to map which regions are essential for immune antagonism

  • Perform co-immunoprecipitation experiments to identify host factors interacting with the 633-659 region

  • Use reporter cell lines expressing RIG-I, MDA5, or TLR3 signaling components to measure interference with immune activation

  • Compare immunomodulatory effects between recombinant 633-659 peptides and full-length ORF2

These approaches will help determine whether the 633-659 region contributes to the immune evasion properties of ORF2.

How does the ORF2 (633-659 a.a.) region contribute to viral capsid assembly and stability?

The contribution of the ORF2 (633-659 a.a.) region to viral capsid assembly requires sophisticated structural analysis. The 633-659 region lies outside the core capsid-forming domain (residues 105-458 of ORF2) , suggesting it may have specialized functions beyond basic capsid formation.

A study of ribavirin-induced ORF2 single-nucleotide variants revealed that mutations in ORF2, particularly P79S, can cause defective, smaller HEV particles with reduced infectiousness . While this mutation is outside the 633-659 region, it demonstrates how alterations in ORF2 can dramatically affect particle morphology and function.

Methodologically, researchers investigating this region's contribution to capsid structure should:

  • Use cryo-electron microscopy to visualize particles formed with and without the 633-659 region

  • Perform thermal stability assays to determine if this region contributes to capsid stability

  • Create recombinant virus-like particles with deletions or mutations in this region to assess assembly efficiency

  • Use atomic force microscopy to measure mechanical properties of capsids with modified 633-659 regions

These approaches will help elucidate whether this region provides structural stability, contributes to receptor binding, or serves primarily as an external immunogenic domain.

What are the optimal expression systems for producing recombinant HEV ORF2 (633-659 a.a.) for research purposes?

The expression system selection for recombinant HEV ORF2 (633-659 a.a.) significantly impacts protein quality and experimental applications. E. coli remains the predominant system, with the protein typically fused with beta-galactosidase at the N-terminus to enhance solubility and expression levels .

Expression SystemAdvantagesLimitationsApplications
E. coliHigh yield, cost-effective, simple purificationLacks glycosylation, potential misfoldingELISA, Western blots, antibody production
Baculovirus/insect cellsProper folding, some PTMs, higher purityModerate yield, higher cost, complex glycans differ from mammalianStructural studies, functional assays
Mammalian cellsNative-like glycosylation, proper foldingLower yield, highest cost, complex purificationNeutralization assays, receptor binding studies
Cell-free systemsRapid production, avoids toxicity issuesLimited PTMs, higher cost, lower yieldEpitope mapping, preliminary screening

Methodology recommendations include:

  • Using E. coli for applications where glycosylation is not required

  • Employing insect or mammalian cells when studying interactions requiring native protein conformation

  • Optimizing purification with proprietary chromatographic techniques to achieve >95% purity

  • Verifying protein quality by circular dichroism spectroscopy to confirm secondary structure

The choice should be guided by the specific research question, as expression system selection directly impacts protein characteristics and experimental outcomes.

How can researchers effectively design ELISA assays using HEV ORF2 (633-659 a.a.) for detecting anti-HEV antibodies?

Designing effective ELISA assays using HEV ORF2 (633-659 a.a.) requires careful optimization of multiple parameters. This region is particularly valuable for HEV detection due to its immunodominance and cross-reactivity with sera from patients infected with different HEV genotypes .

A methodical approach to ELISA development should include:

  • Optimization of coating conditions:

    • Buffer composition: Carbonate buffer (pH 9.6) typically yields optimal protein binding

    • Protein concentration: Titrate between 0.5-5 μg/ml to determine optimal signal-to-noise ratio

    • Incubation time: Compare overnight at 4°C versus 2 hours at 37°C

  • Blocking and sample dilution:

    • Test different blocking agents (BSA, casein, or commercial blockers)

    • Optimize serum dilutions (typically 1:100 to 1:1000) to minimize background

    • Include genotype-specific positive controls when possible

  • Detection system selection:

    • HRP-conjugated secondary antibodies with TMB substrate provide excellent sensitivity

    • Consider biotinylated proteins with streptavidin-HRP for enhanced signal amplification

    • Evaluate colorimetric versus chemiluminescent detection based on sensitivity requirements

  • Validation parameters:

    • Determine sensitivity and specificity using well-characterized serum panels

    • Establish cut-off values through ROC curve analysis

    • Perform cross-reactivity testing with other hepatitis viruses (HAV, HBV, HCV)

Researchers should note that the immunoreactivity of this region with sera from HEV-infected individuals makes it particularly suitable for diagnostic applications with minimal specificity problems .

What approaches can be used to study the interaction between HEV ORF2 (633-659 a.a.) and neutralizing antibodies?

Investigating interactions between HEV ORF2 (633-659 a.a.) and neutralizing antibodies requires sophisticated methodologies that capture both binding kinetics and functional outcomes. Recent research indicates that secreted forms of ORF2 can interfere with antibody-mediated neutralization, suggesting complex antibody-antigen dynamics .

Advanced experimental approaches include:

  • Epitope mapping techniques:

    • Peptide arrays covering the 633-659 region with single amino acid resolution

    • Hydrogen-deuterium exchange mass spectrometry to identify antibody footprints

    • Alanine scanning mutagenesis to identify critical binding residues

  • Binding kinetics assessment:

    • Surface plasmon resonance (SPR) to determine kon and koff rates

    • Bio-layer interferometry for real-time, label-free analysis

    • Isothermal titration calorimetry to characterize thermodynamic parameters

  • Neutralization interference assays:

    • Competition assays with soluble ORF2 and infectious HEV particles

    • Preincubation studies to assess antibody blocking by secreted ORF2 forms

    • Flow cytometry to measure inhibition of virus attachment to susceptible cells

  • Structural analysis of antibody-antigen complexes:

    • X-ray crystallography of Fab fragments bound to ORF2 peptides

    • Cryo-electron microscopy of neutralizing antibodies bound to virus-like particles

    • Computational docking and molecular dynamics simulations

These approaches can reveal whether antibodies targeting the 633-659 region neutralize the virus by blocking receptor binding sites, preventing conformational changes, or inducing aggregation of viral particles.

How do single nucleotide variants in the ORF2 region affect HEV infectivity and immune evasion?

Single nucleotide variants (SNVs) in the ORF2 region can significantly impact HEV biology, as demonstrated by studies on ribavirin-induced mutations. Research shows that certain ORF2 variants, particularly P79S, demonstrate reduced RNA copy numbers in supernatants and impaired production of infectious particles . These variants produce defective, smaller HEV particles with compromised infectiousness .

The implications extend beyond structural changes to immune interactions. The P79S variant displays altered subcellular distribution of the ORF2 protein and can interfere with antibody-mediated neutralization of HEV, potentially acting as an immune decoy . This suggests that naturally occurring or treatment-induced SNVs may contribute to viral immune evasion strategies.

Methodologically, researchers investigating SNVs should:

  • Use site-directed mutagenesis to introduce specific variants into infectious clones

  • Employ next-generation sequencing to identify minor variant populations in clinical samples

  • Develop competition assays to assess how variants affect neutralization by monoclonal or polyclonal antibodies

  • Perform longitudinal studies in chronic HEV patients to track the emergence of variants during treatment

This research direction offers opportunities to understand viral evolution during persistent infection and may guide personalized antiviral strategies in the future.

What is the distinct role of secreted forms of ORF2 (ORF2s) versus capsid-associated ORF2 in HEV pathogenesis?

Recent research has elucidated distinct forms of the ORF2 protein with separate origins and functions. The secreted form of ORF2 (ORF2s) and the capsid form (ORF2c) are different translation products . ORF2s results from translation initiated at the previously presumed AUG start codon, while ORF2c originates from a previously unrecognized internal AUG codon (15 codons downstream) .

The additional 15 amino acids in ORF2s create a signal sequence driving secretion via the secretory pathway . Unlike ORF2c, ORF2s is glycosylated and exists as a dimer . While ORF2s exhibits substantial antigenic overlap with the capsid, epitopes predicted to bind the putative cell receptor are lost, meaning ORF2s does not block HEV cell entry but inhibits antibody-mediated neutralization .

In vivo studies suggest ORF2s may be dispensable for viral replication but is required for long-lived antibody-mediated responses that protect against infection . Furthermore, serum ORF2s levels increase during persistent infection compared to acute infection , suggesting a role in establishing chronicity.

Researchers investigating these distinct forms should:

  • Develop systems to separately track and quantify ORF2s and ORF2c in patient samples

  • Create viral mutants that selectively express only one form to assess their individual contributions

  • Examine the interplay between ORF2s and host immune components, particularly neutralizing antibodies

  • Evaluate ORF2s as a potential biomarker for prediction of chronic versus self-limited infection

These approaches will help clarify the complex roles of different ORF2 forms in HEV pathogenesis and persistence.

How can structural knowledge of the ORF2 (633-659 a.a.) region be leveraged for developing improved HEV vaccines?

Current understanding of the ORF2 (633-659 a.a.) immunodominant region offers opportunities for rational vaccine design. This region contains important B-cell epitopes that elicit neutralizing antibodies, making it a valuable component for subunit vaccine approaches.

Several strategic considerations for vaccine development include:

  • Epitope optimization strategies:

    • Consensus sequence design based on multiple genotypes to provide broader protection

    • Multimeric display on virus-like particles to enhance immunogenicity

    • Strategic mutation of non-essential residues to focus immune responses on conserved neutralizing epitopes

  • Delivery platform evaluation:

    • Recombinant protein formulations with modern adjuvants

    • mRNA vaccines encoding optimized ORF2 constructs

    • Viral vector vaccines (adenovirus or VSV) expressing ORF2 regions

  • Immune response assessment metrics:

    • Neutralizing antibody titers against multiple genotypes

    • Duration of antibody responses (6-12 months minimum)

    • T-cell responses to complementary epitopes within ORF2

    • Protection against challenge in appropriate animal models

  • Special population considerations:

    • Enhanced immunogenicity formulations for immunocompromised patients

    • Dose optimization for pregnant women (a high-risk group for severe HEV)

    • Age-appropriate formulations for pediatric and elderly populations

The methodological approach should include comparative studies of candidates expressing different ORF2 regions to determine whether the 633-659 a.a. region alone is sufficient or if additional epitopes are required for robust protection against diverse HEV strains.

What are the conflicting reports regarding ORF2 protein translation and processing?

A significant controversy exists regarding the origin and processing of different ORF2 forms. Conflicting reports indicate that secreted ORF2 (ORF2s) and capsid ORF2 (ORF2c) are generated either by:

  • Translation initiation at the same start codon with subsequent proteolytic cleavage, or

  • Translation initiation at different start codons, leading to a signal peptide on ORF2s and differential cellular localization and processing .

One study provided evidence that production of ORF2s results from translation initiated at the previously presumed AUG start codon for the capsid protein, whereas translation of the actual capsid protein (ORF2c) is initiated at a previously unrecognized internal AUG codon (15 codons downstream) . The addition of these 15 amino acids creates a signal sequence driving ORF2s secretion .

Methodologically, researchers addressing this controversy should:

  • Perform ribosome profiling to identify translation initiation sites in infected cells

  • Use CRISPR-Cas9 to mutate potential start codons in infectious clones

  • Employ pulse-chase experiments with specific antibodies to track protein processing

  • Analyze leader sequences using signal peptide prediction algorithms and validate experimentally

Resolving this controversy is crucial for understanding ORF2 biology and developing interventions targeting specific ORF2 forms.

How reliable are current diagnostic assays based on recombinant ORF2 (633-659 a.a.) compared to full-length ORF2?

The reliability of diagnostic assays using the ORF2 (633-659 a.a.) fragment versus full-length ORF2 presents important considerations for clinical diagnostics. While the 633-659 region is immunodominant and suitable for HEV detection with "minimal specificity problems" , potential limitations exist.

Comparative analysis considerations:

ParameterORF2 (633-659 a.a.)Full-length ORF2Implications
SensitivityGood for detecting strong antibody responsesMay detect broader range of antibody specificitiesPotential false negatives with fragment-based assays in early infection
SpecificityHigh due to immunodominant epitopesVariable depending on cross-reactive epitopesFragment may offer better specificity for confirmatory testing
Cross-genotype detectionGood for conserved epitopesBetter for capturing genotype-specific responsesFragment-based assays may miss genotype-specific antibodies
Production complexitySimpler, higher yieldMore complex expression, lower yieldFragment-based tests may be more cost-effective
Conformational epitopesLimited representationBetter representationMay miss antibodies targeting discontinuous epitopes

Researchers evaluating these assays should:

  • Perform head-to-head comparisons using well-characterized serum panels

  • Assess performance across different disease stages (acute, convalescent, chronic)

  • Evaluate cross-reactivity with antibodies against other hepatitis viruses

  • Consider combining multiple ORF2 regions for optimal performance

Understanding these trade-offs is essential for selecting appropriate diagnostic tools for different clinical and research applications.

What are the challenges in translating in vitro findings about ORF2 (633-659 a.a.) function to in vivo infection models?

Translating in vitro findings about HEV ORF2 (633-659 a.a.) to in vivo infection models presents several methodological challenges that researchers must address. These challenges stem from differences between controlled laboratory conditions and the complex environment of living organisms.

Key translational challenges include:

  • Model system limitations:

    • Limited availability of permissive animal models that recapitulate human HEV infection

    • Differences in immune responses between species that may affect ORF2 interactions

    • Variations in receptor distribution and tissue tropism affecting ORF2 functionality

  • Experimental design considerations:

    • Difficulty in delivering recombinant proteins to relevant tissues in vivo

    • Challenges in distinguishing between effects of different ORF2 forms in vivo

    • Limited ability to perform high-resolution imaging of ORF2 localization in intact tissues

  • Technical methodological approaches:

    • Development of genetically modified HEV strains that express tagged ORF2 variants

    • Utilization of humanized mouse models expressing human factors necessary for HEV infection

    • Employment of in situ hybridization and immunohistochemistry to track viral components

    • Application of advanced tissue clearing and 3D imaging techniques

  • Data interpretation complexities:

    • Distinguishing direct effects of ORF2 from secondary consequences of infection

    • Accounting for host genetic factors that may influence ORF2 interactions

    • Considering the impact of viral quasi-species diversity on ORF2 function

Addressing these challenges requires interdisciplinary approaches combining molecular virology, immunology, and advanced imaging techniques to bridge the gap between in vitro observations and in vivo relevance.

Product Science Overview

Introduction

Hepatitis E virus (HEV) is a positive-sense, single-stranded RNA virus that belongs to the Hepeviridae family . It is a zoonotic pathogen responsible for causing hepatitis in both humans and animals, leading to acute hepatitis E outbreaks worldwide . The HEV genome is approximately 7.2 kb in length and encodes three well-defined open reading frames (ORFs): ORF1, ORF2, and ORF3 .

ORF2 Protein

The ORF2 protein is the major structural protein of HEV and forms the viral capsid . It plays a crucial role in the virus’s life cycle, including viral replication, pathogenesis, and immune response regulation . The ORF2 protein is also involved in the development of vaccines and diagnostic tools for HEV .

Recombinant ORF2 (633-659 a.a.)

The recombinant ORF2 (633-659 a.a.) is a specific fragment of the ORF2 protein. This fragment has been studied for its potential applications in immunization protocols and antibody generation . The recombinant protein is produced using advanced biotechnological methods to ensure high sensitivity, specificity, and reproducibility .

Biological Properties and Functions

The ORF2 protein, including the 633-659 a.a. fragment, has multiple functions beyond forming the viral capsid. It is involved in:

  • Viral Replication: The ORF2 protein plays a role in the replication of HEV by interacting with host cellular machinery .
  • Immune Response Regulation: The protein modulates the host’s innate immune response and cellular signaling pathways .
  • Pathogenesis: ORF2 is implicated in the pathogenesis of HEV, contributing to the virus’s ability to cause disease .
Applications in Research and Medicine

The recombinant ORF2 (633-659 a.a.) fragment is valuable in various research and medical applications:

  • Vaccine Development: The ORF2 protein is a target for vaccine development due to its immunogenic properties .
  • Diagnostic Tools: Recombinant ORF2 fragments are used in diagnostic assays to detect HEV infections .
  • Antibody Generation: The recombinant protein is used to generate specific antibodies for research and therapeutic purposes .

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.