HAV P3C

Hepatitis A Virus P3C Recombinant
Shipped with Ice Packs
In Stock

Description

Production and Stability

HAV P3C is recombinantly produced in E. coli and undergoes rigorous purification to ensure high specificity. Stability and handling guidelines include:

ParameterSpecificationSource
Storage Temperature≤ -18°C (avoid freeze-thaw cycles)
Shelf Life12 months
FormulationLiquid (10 mM CBB, pH 9.6, 0.1% SDS, 50% glycerol)

The protein is stable at 4°C for short-term storage (1 week) but requires cryogenic preservation for extended use .

Applications in Research and Diagnostics

HAV P3C serves as a critical antigen in immunological assays due to its immunodominant regions:

Diagnostic Applications

  • ELISA and Western Blot: HAV P3C is used to detect HAV-specific antibodies in patient sera, with minimal cross-reactivity .

  • Antigenic Profiling: Among 42 antigenic domains identified in the HAV polyprotein, the P3C region (1643–1743) is one of five immunodominant regions, making it a reliable target for serological testing .

Immunogenicity Studies

  • Antibody Reactivity: HAV P3C reacts strongly with sera from HAV-infected individuals, confirming its role in eliciting immune responses .

  • Antigenic Domains: The C-terminal region of P3C (1643–1743) overlaps with the P3D protein, forming a junctional domain recognized by host immune systems .

Therapeutic Relevance

While HAV P3C is not a direct therapeutic target, its structural homology to other viral proteins informs antiviral strategies. For example:

  • Inhibitor Design: Studies on HAV 3C protease (e.g., phthalhydrazine-based inhibitors) highlight the importance of targeting enzymatic cleavage sites in polyprotein processing .

  • Combination Therapy: Inhibitors of HAV 3C, such as Z10325150, show synergy with RNA-dependent RNA polymerase inhibitors (e.g., favipiravir) in reducing viral replication .

Product Specs

Introduction
Forty-two antigenic domains of the hepatitis A virus (HAV) polyprotein were identified using 237 overlapping synthetic peptides (20-mers) that spanned the entire HAV polyprotein. Nineteen antigenic domains were located within the structural proteins, and 22 were found within the nonstructural proteins. One domain spanned the junction of the VP1 and P2A proteins. Five domains were classified as immunodominant based on their breadth and strength of immunoreactivity. One domain is located within the VP2 protein at amino acid positions 57-90. A second domain, located at positions 767-842, contains the C-terminal region of the VP1 protein and the complete P2A protein. A third domain, located at positions 1403-1456, includes the C-terminal portion of the P2C protein and the N-terminal half of the P3A protein. The fourth domain, located at positions 1500-1519, encompasses almost the entire P3B protein. The final domain, located at positions 1719-1764, contains the C-terminal region of the P3C protein and the N-terminal region of the P3D protein. Notably, four of the five most immunoreactive domains originate from small HAV proteins and/or span protein cleavage sites between different HAV proteins.
Description
This recombinant protein is derived from E. coli and has a molecular weight of 40 kDa. It contains the immunodominant regions of the HAV P3C protein, specifically amino acids 1643-1743.
Purity
The purity of the HAV P3C protein is greater than 90%, as determined by 10% SDS-PAGE and Coomassie blue staining.
Formulation
The HAV P3C protein is supplied in a buffer consisting of 10mM CBB (Coomassie Brilliant Blue), pH 9.6, 0.1% SDS (sodium dodecyl sulfate), and 50% glycerol.
Stability
The HAV P3C protein is stable at 4°C for 1 week. However, for long-term storage, it is recommended to store the protein below -18°C. Avoid repeated freeze-thaw cycles.
Applications
The HAV P3C antigen is suitable for use in enzyme-linked immunosorbent assays (ELISA) and Western blots. It serves as an excellent antigen for the detection of HAV with minimal specificity issues.
Source
Escherichia Coli.
Purification Method
HAV P3C protein was purified by proprietary chromatographic technique.
Specificity
Immunoreactive with sera HAV-infected individuals.

Q&A

What is the structural composition of HAV 3C protease?

HAV 3C protease contains a catalytic triad consisting of His44, Asp84, and Cys172, which forms the active site crucial for its proteolytic function. Crystallographic studies have revealed that these residues create a catalytic site that serves as a useful target for protease-inhibitor interactions. The protein structure has been determined via X-ray crystallography, providing essential structural information for molecular docking studies and drug development efforts .

How does HAV 3C protease contribute to viral replication?

HAV 3C protease plays an essential role in viral polyprotein processing, which is critical for HAV replication and infection. The enzyme cleaves the HAV polyprotein at specific junctions, generating functional individual viral proteins required for genome replication and virion assembly. Studies have demonstrated that inhibition of this protease significantly reduces HAV replication, confirming its critical role in the viral life cycle . The process involves both intramolecular (in cis) and potentially intermolecular (in trans) cleavage reactions, with evidence suggesting that the P2-P3 junction cleavage occurs primarily through intramolecular reactions .

What substrate specificity does HAV 3C protease exhibit?

HAV 3C protease shows distinct amino acid preferences at its cleavage sites. Research using a rapid screening method with N-terminal acetylated peptide mixtures revealed that the enzyme prefers glycine, alanine, and serine at the P'1 position. At the P'2 position, the enzyme exhibits limited specificity, excluding only arginine and proline peptides as substrates. This substrate specificity profile differs somewhat from other picornavirus 3C proteases, potentially offering opportunities for selective inhibitor design .

PositionPreferred Amino AcidsExcluded Amino Acids
P'1Glycine, Alanine, SerineMost others (lower preference)
P'2Most amino acids (limited specificity)Arginine, Proline

What computational methods are effective for screening potential HAV 3C protease inhibitors?

Computational screening for HAV 3C protease inhibitors effectively uses molecular docking approaches to identify potential inhibitory compounds. The Schrödinger Glide program has been successfully employed for this purpose, positioning three-dimensional models of small molecules into the HAV 3C binding pocket and predicting binding affinities through associated scoring functions. The process typically includes:

  • Structure preparation of proteins and ligands using Schrödinger Maestro

  • Generation of a grid near the catalytic triad (His44, Asp84, and Cys172)

  • Molecular docking simulations using the Schrödinger Glide program

  • Selection of compounds based on Glide scores and binding interactions

Researchers should note that while docking scores provide useful guidance, they aren't always directly correlated with biological activity. The selection of compounds should consider both docking scores and specific functional group interactions within the active site .

How can researchers validate in silico findings through in vitro studies for HAV 3C inhibitors?

Validation of in silico predictions requires comprehensive in vitro testing protocols. Based on successful research approaches, the following methodology is recommended:

  • Select lead compounds from in silico screening based on docking scores and predicted interactions

  • Evaluate inhibitory effects on HAV replication using appropriate cell culture systems:

    • HAV genotype IB subgenomic replicon in HuhT7 cells

    • HAV genotype IIIA HA11-1299 in Huh7 cells

  • Measure viral replication through quantitative techniques (e.g., RT-qPCR)

  • Assess cytotoxicity of compounds at various concentrations

  • Determine the efficacy of lead compounds alone and in combination therapy

This approach has proven effective in identifying compounds such as Z10325150, which demonstrated significant inhibitory effects on HAV replication without cytotoxicity at concentrations up to 100 μg/mL .

What experimental approaches can determine the substrate specificity of HAV 3C protease?

The substrate specificity of HAV 3C protease can be determined through a rapid screening method using peptide mixtures. The protocol involves:

  • Prepare N-terminal acetylated peptide mixtures identical in sequence except for positions of interest (e.g., P'1 or P'2)

  • Introduce a set of 15-16 amino acids at these positions

  • Expose the peptide mixtures to HAV 3C protease

  • Analyze enzyme-catalyzed hydrolysis products by Edman degradation

  • Calculate the relative yield of each amino acid product to determine relative kcat/Km values

This method provides simultaneous evaluation of multiple potential substrates and efficiently identifies preferred residues for peptide substrates. The approach is applicable to other endoproteinases and offers a significant advantage over testing individual peptides sequentially .

What characteristics make HAV 3C protease a viable target for antiviral drug development?

HAV 3C protease presents several characteristics that make it an attractive target for antiviral drug development:

  • Essential role in viral replication: 3C protease is critical for polyprotein processing and promotion of HAV replication

  • Conserved structure: The catalytic site formed by the triad His44, Asp84, and Cys172 is well-characterized and conserved

  • Success with related viruses: Protease inhibitors have played important roles in treating other viral infections like HIV-1 and HCV

  • Demonstrated inhibition: Compounds targeting this protease have shown significant inhibition of HAV replication

  • Cross-genotype activity: Some inhibitors (like Z10325150) have demonstrated efficacy against multiple HAV genotypes (IB and IIIA)

The success of protease inhibitors for other viral infections provides a compelling rationale for pursuing HAV 3C protease as a therapeutic target, with evidence already demonstrating that compounds binding to the active site can inhibit viral replication .

How does combination therapy with HAV 3C protease inhibitors compare to monotherapy?

Combination therapy with HAV 3C protease inhibitors and other antiviral agents demonstrates enhanced efficacy compared to monotherapy. Research shows that combining Z10325150 (a HAV 3C protease inhibitor) with favipiravir (an RNA-dependent RNA polymerase inhibitor) produces significantly improved inhibition of HAV replication:

Treatment ApproachInhibition of HAV IB HM175 Subgenomic RepliconInhibition of HAV IIIA HA11-1299 Replication
Z10325150 Monotherapy47%36%
Z10325150 + Favipiravir64%48%

This synergistic effect highlights the potential of targeting multiple viral replication mechanisms simultaneously, a strategy that has proven successful with other viral infections. The combination approach may offer advantages in terms of enhanced efficacy, potential dose reduction, and reduced likelihood of resistance development .

What structural features of inhibitor compounds correlate with effective HAV 3C protease inhibition?

Key structural features that correlate with effective HAV 3C protease inhibition include:

It's worth noting that compounds with extremely high docking scores don't necessarily exhibit the best biological activity, emphasizing the importance of analyzing specific interactions within the active site rather than relying solely on computational scores .

How does the mechanism of HAV 3C protease differ from other picornavirus 3C proteases?

HAV 3C protease exhibits distinct differences from other picornavirus 3C proteases in several key aspects:

  • Substrate specificity: HAV 3C shows preference for glycine, alanine, and serine at the P'1 position, while having limited specificity at the P'2 position (excluding only arginine and proline). This profile differs from other picornavirus 3C proteases

  • Cleavage mechanisms: HAV 3C appears to favor intramolecular (cis) cleavage at the P2-P3 junction, with experiments suggesting difficulties in demonstrating intermolecular cutting of this junction

  • Antigenic properties: HAV 3C contains antigenic epitopes that can be efficiently modeled with short synthetic peptides, particularly at protein cleavage sites separating different HAV proteins

  • Immunoreactivity profile: Antibodies to P3C protein are detected in experimentally infected primates and acutely infected patients, but not in primates immunized with inactivated HAV

These differences provide opportunities for developing HAV-specific inhibitors and diagnostic tools that differentiate between inactivated vaccine-induced immunity and natural infection .

What is known about the temporal sequence of polyprotein processing by HAV 3C protease?

The temporal sequence of HAV polyprotein processing by 3C protease follows a specific order, as revealed by pulse-chase experiments:

  • Initial rapid cleavage: The P2-P3 junction undergoes fast cleavage, representing the first step in polyprotein processing

  • Secondary processing: Further but incomplete processing occurs at the 3C-3D junction

  • Mechanism dependency: Mutation of the 3C coding sequence eliminates all cleavages, confirming the critical role of 3C protease activity

  • Processing directionality: The cleavage at the P2-P3 junction likely occurs through intramolecular reactions (in cis), as efforts to demonstrate intermolecular cutting of this junction by active 3C or 3CD sequences were unsuccessful

This ordered processing ensures the correct generation of functional viral proteins required for replication. The incomplete processing at certain junctions may represent a regulatory mechanism to maintain optimal ratios of viral proteins during different stages of infection .

How can antigenic epitopes within HAV 3C protein be utilized for diagnostic or research applications?

Antigenic epitopes within HAV 3C protein offer valuable applications in diagnostics and research:

  • Differential diagnosis: HAV 3C antibodies appear uniquely in cases of natural infection but not in vaccinated individuals, offering a way to differentiate between vaccine-induced immunity and natural infection

  • Infection markers: Antibodies to nonstructural proteins like P3C in patient serum serve as markers of active viral replication

  • Epitope mapping: Studies have identified specific antigenic domains within HAV proteins, including the C-terminal region of P3C protein and the N-terminal region of P3D protein (position 1719-1764 aa)

  • Synthetic peptide models: Short synthetic peptides can efficiently model these antigenic epitopes, providing tools for antibody detection and characterization

Research has demonstrated that some of the most immunoreactive domains are derived from small HAV proteins and/or encompass protein cleavage sites separating different HAV proteins. The ability to model these epitopes with synthetic peptides provides opportunities for improved diagnostic assays and better understanding of the immune response to HAV infection .

What are the major limitations in current HAV 3C protease research methodologies?

Current HAV 3C protease research faces several methodological limitations:

  • Molecular docking inaccuracies: Computational methods for predicting binding interactions have inherent inaccuracies that can lead to false positives or missed opportunities

  • Limited correlation between docking scores and biological activity: High docking scores don't always translate to effective biological activity, requiring additional validation

  • Cell culture system constraints: HAV grows poorly in cell culture compared to other picornaviruses, making in vitro evaluation of inhibitors challenging

  • Cross-genotype efficacy: Differential effectiveness against different HAV genotypes complicates the development of broadly effective inhibitors

  • Translation to in vivo models: Limited animal models for HAV infection make it difficult to validate findings from in vitro and in silico studies

Researchers should be aware of these limitations when designing experiments and interpreting results, particularly noting that molecular docking studies should be complemented with functional validation and that compounds may show variable efficacy across different HAV genotypes .

How can emerging technologies advance our understanding of HAV 3C protease function and inhibition?

Emerging technologies offer promising avenues to advance HAV 3C protease research:

  • Cryo-electron microscopy: High-resolution structural analysis of HAV 3C protease and its complexes with inhibitors or substrates in near-native states

  • Molecular dynamics simulations: Enhanced computational approaches to better predict protein-ligand interactions and account for protein flexibility

  • Fragment-based drug discovery: Identification of small molecular fragments that bind to HAV 3C, which can be optimized into more potent inhibitors

  • Proteomics approaches: Mass spectrometry-based techniques to comprehensively identify all cleavage sites and better understand substrate specificity

  • CRISPR-Cas9 gene editing: Creating cell lines with modifications to host factors that interact with HAV 3C to understand the broader role of this protease in the cellular context

These technologies can overcome current limitations in structural analysis, provide more accurate predictions of binding interactions, and enable a systems-level understanding of HAV 3C protease in the viral life cycle .

What are the prospects for developing broad-spectrum inhibitors targeting multiple picornavirus 3C proteases?

The development of broad-spectrum inhibitors targeting multiple picornavirus 3C proteases shows promising potential:

Product Science Overview

Introduction

Hepatitis A virus (HAV) is a member of the Picornaviridae family and is responsible for causing acute hepatitis in humans. The virus has a positive-sense single-stranded RNA genome of approximately 7.5 kb, encapsidated in a protein shell composed of viral proteins VP1, VP2, and VP3 . The P3C protein, also known as 3C protease, plays a crucial role in the viral life cycle by processing the viral polyprotein and interacting with host cell proteins .

Structure and Function of P3C

The P3C protease is a cysteine protease that cleaves the viral polyprotein at specific sites to generate functional viral proteins. This protease is essential for the maturation of the viral particles and the replication of the virus . In addition to its role in polyprotein processing, P3C protease also interacts with host cell proteins to modulate the host’s antiviral response, transcription, and translation processes .

Recombinant P3C Protein

Recombinant P3C protein refers to the P3C protease that has been produced using recombinant DNA technology. This involves cloning the gene encoding the P3C protease into an expression vector, which is then introduced into a host cell (such as E. coli) to produce the protein in large quantities. Recombinant P3C protein is used in various research applications, including studies on viral replication, protein-protein interactions, and the development of antiviral drugs.

Applications in Research
  1. Viral Replication Studies: Recombinant P3C protein is used to study the mechanisms of HAV replication and the role of P3C protease in the viral life cycle. By understanding how P3C protease processes the viral polyprotein, researchers can identify potential targets for antiviral drugs .
  2. Protein-Protein Interactions: The interaction of P3C protease with host cell proteins is crucial for the virus to evade the host’s immune response. Recombinant P3C protein is used to identify and characterize these interactions, providing insights into the virus-host relationship .
  3. Antiviral Drug Development: Inhibitors of P3C protease are potential candidates for antiviral drugs. Recombinant P3C protein is used in high-throughput screening assays to identify compounds that can inhibit the protease activity, thereby blocking viral replication .

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.