HCV NS3, Biotin

Hepatitis C Virus NS3, Biotin Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to HCV NS3 Protein

Hepatitis C virus (HCV) non-structural protein 3 (NS3) is a bifunctional enzyme critical for viral replication. It comprises an N-terminal serine protease domain and a C-terminal RNA helicase domain . The protease cleaves the viral polyprotein to release non-structural proteins , while the helicase unwinds RNA during replication . NS3 also disrupts host immune responses by targeting mitochondrial antiviral signaling proteins . Due to its essential role, NS3 is a key target for diagnostic and therapeutic research .

Biotinylated HCV NS3 (HCV NS3, Biotin) is a recombinant protein engineered for enhanced detection in assays. It is widely used in research to study HCV pathogenesis and develop antiviral strategies .

Protease Domain

  • Structure: The NS3 protease adopts a chymotrypsin-like fold with two β-barrel domains and a catalytic triad (Ser-139, His-57, Asp-81) . NS4A cofactor binding stabilizes the active site and improves substrate specificity .

  • Substrate Recognition: Cleaves polyprotein junctions (e.g., NS3-NS4A, NS4B-NS5A) and host immune proteins .

Helicase Domain

  • Mechanism: Uses ATP hydrolysis to translocate along RNA/DNA via an "inchworm" model, involving conformational shifts between domains 1 and 2 .

  • Conserved Motifs: Contains helicase motifs critical for ATP and nucleic acid binding .

Immunoassays

  • Antigen Detection: HCV NS3, Biotin serves as a high-specificity antigen for detecting anti-HCV antibodies in patient sera .

  • Binding Studies: Used to map antibody-epitope interactions or evaluate inhibitor binding kinetics .

Mechanistic Studies

  • Fibrosis Research: NS3 mimics TGF-β2 by binding TGF-β receptor I (TβRI), promoting liver fibrosis . Biotinylated NS3 enables tracking of these interactions via streptavidin-based assays.

  • Drug Resistance: Structural studies using labeled NS3 reveal how mutations (e.g., A156T, D168V) reduce inhibitor efficacy .

Stability and Handling Protocols

  • Short-Term Storage: Stable at 4°C for 1 week .

  • Long-Term Storage: Below -18°C in urea-containing buffer to prevent aggregation .

  • Buffer Optimization: β-mercaptoethanol maintains protein solubility and reduces disulfide bond formation .

Protease Inhibitors

  • First-Generation Inhibitors: Telaprevir and ciluprevir target the NS3 active site but face resistance due to mutations outside the substrate envelope .

  • Next-Generation Designs: Macrocyclic inhibitors (e.g., TMC-435) exploit interactions with helicase residues to improve efficacy .

Helicase Inhibitors

  • Peptide Inhibitors: The p14 peptide inhibits helicase activity (IC50 = 725 nM) by binding domain 1 .

  • Therapeutic Potential: Blocking helicase activity disrupts viral replication without affecting host enzymes .

Product Specs

Introduction
Hepatitis C virus (HCV) is a small (50nm), enveloped virus with a single-stranded, positive-sense RNA genome. It belongs to the family Flaviviridae. HCV exhibits a rapid replication rate, producing roughly one trillion particles daily within an infected individual. The virus's RNA polymerase lacks proofreading ability, resulting in a high mutation rate, which contributes to its ability to evade the host's immune system. HCV is categorized into six genotypes (1-6), each with multiple subtypes. The prevalence and distribution of these genotypes differ globally. Genotype determination is clinically crucial as it influences the potential response to interferon-based therapy and the required treatment duration. Genotypes 1 and 4 demonstrate lower responsiveness to interferon-based treatment compared to genotypes 2, 3, 5, and 6.
Description
This product consists of a recombinant HCV NS3 protein, produced in E.coli, containing the immunodominant regions of the HCV NS3 protein (amino acids 1450-1643), resulting in a 22 kDa protein. This protein is biotinylated and features a 6xHis-Tag at the N-terminus.
Purity
The purity of the HCV NS3 Biotinylated protein exceeds 95%, as determined by 10% SDS-PAGE analysis with Coomassie blue staining.
Formulation
The protein is supplied in a solution containing 1.5M urea, 20mM Tris-HCl (pH 8.0), and 10mM β-mercaptoethanol at a concentration of 1mg/ml.
Stability
While the HCV NS3 Biotinylated protein remains stable for 1 week when stored at 4°C, long-term storage at or below -18°C is recommended. Repeated freeze-thaw cycles should be avoided.
Applications
The HCV NS3 Biotinylated antigen is suitable for use in various applications, including ELISA and Western blots. It serves as an excellent antigen for the detection of HCV with minimal specificity issues.
Purification Method
HCV NS3 Biotin labeled protein was purified by proprietary chromatographic technique.
Specificity
Reactivity with human HCV positive serum undetermined.

Q&A

What is HCV NS3 and why is it important in HCV research?

HCV NS3 is a multifunctional viral protein that plays crucial roles in the hepatitis C virus life cycle. It possesses three distinct enzymatic activities: serine protease (N-terminal domain), RNA helicase, and NTPase (C-terminal domain). These enzymatic functions are essential for viral replication, as NS3 processes the viral polyprotein and facilitates RNA replication through its helicase activity. Its critical role makes NS3 an important target for antiviral drug development and basic research into HCV pathogenesis .

HCV belongs to the Flaviviridae family and is classified into at least six major genotypes (1-6) with several subtypes. The virus demonstrates an exceptionally high replication rate, producing approximately one trillion particles daily in infected individuals. This high replication rate, combined with the lack of proofreading by HCV RNA polymerase, results in an unusually high mutation rate that helps the virus evade host immune responses .

How is biotin-labeled HCV NS3 protein produced and purified for research applications?

Biotin-labeled HCV NS3 protein is typically produced through recombinant expression systems, with E. coli being the most common platform. The production process involves several key steps:

  • Gene cloning: The HCV NS3 gene sequence (typically containing immunodominant regions, a.a. 1450-1643) is cloned into an expression vector with a 6xHis-Tag at the N-terminus to facilitate purification.

  • Expression: Recombinant protein is expressed in bacterial systems under optimized conditions.

  • Biotinylation: Site-specific biotinylation is performed, often at the N-terminus to minimize interference with the protein's functional domains.

  • Purification: The biotin-labeled protein is purified using proprietary chromatographic techniques to achieve high purity (>95% as determined by PAGE) .

The resulting protein typically has a molecular weight of approximately 22 kDa and contains the immunodominant regions of the HCV NS3 helicase domain. Quality control includes verification of biotin labeling efficiency and retention of enzymatic activities .

What are the functional differences between NS3 proteins from different HCV genotypes?

HCV is classified into multiple genotypes (1-11) with genotypes 1-6 being the major ones. NS3 proteins from different genotypes exhibit significant sequence variations that affect:

  • Antibody recognition: Monoclonal antibodies may recognize specific genotypes but not others. For example, the antibody clone 1878 recognizes HCV genotypes 1a, 1b, and 2c but not 2a .

  • Treatment response: Genotypes 1 and 4 are generally less responsive to interferon-based treatments compared to genotypes 2, 3, 5, and 6 .

  • Enzyme kinetics: Different genotypes may display variations in protease and helicase activities, affecting replication efficiency and potentially drug susceptibility.

  • Structural epitopes: Conformational differences between genotypes can impact protein-protein interactions and antibody binding sites.

These variations have significant implications for diagnostic test development, therapeutic approaches, and experimental design. Researchers should carefully select genotype-specific or pan-genotypic NS3 reagents based on their experimental goals .

How can biotin-labeled NS3 be used to study NS3-mediated inhibition of HCV replication?

Biotin-labeled NS3 provides a valuable tool for studying inhibition mechanisms of HCV replication through several methodological approaches:

  • Cell-free helicase assays: Biotin-labeled NS3 can be immobilized on streptavidin surfaces to screen for compounds that inhibit its helicase activity. The specificity of inhibition can be confirmed by:

    • Performing reactions in the absence of ATP to verify ATP-dependence

    • Using increasing concentrations of inhibitory compounds

    • Including appropriate controls (unrelated antibodies or known NS3 inhibitors)

  • Cell-based replication models:

    • Replicon systems: Stable cell lines replicating subgenomic RNA can be transfected with anti-NS3 antibodies or inhibitors

    • Transient replication models: Full-length HCV RNA can be used to assess inhibitor effects on viral replication

    • Measurement endpoints: HCV RNA levels (via RT-PCR), viral protein expression (via immunostaining), and negative-strand RNA synthesis (via ribonuclease protection assays)

  • Protein-protein interaction studies:

    • Biotin-labeled NS3 can be used to identify and characterize host factors that interact with NS3

    • Pull-down assays followed by mass spectrometry can reveal novel interaction partners

    • These interactions can be targeted for therapeutic intervention

Experimental data shows that anti-NS3 antibodies can completely inhibit helicase activity at equimolar concentrations in cell-free systems, while intracellular expression of anti-NS3 antibodies results in significant reduction of HCV RNA and viral protein expression in cellular models .

What methods are available for studying the role of NS3 in liver fibrosis development?

HCV NS3 has been implicated in liver fibrosis pathogenesis through mechanisms independent of its enzymatic activities. The following methodological approaches can be used to study this role:

  • Receptor binding studies:

    • Surface Plasmon Resonance (SPR) with immobilized biotin-NS3 can measure binding kinetics to TGF-β receptors

    • Competitive binding assays with TGF-β2 can determine if NS3 mimics TGF-β2 activity

    • Co-immunoprecipitation experiments can verify physical interaction between NS3 and TGF-β receptors in cellular contexts

  • Signaling pathway analysis:

    • Reporter gene assays using (CAGA)9-Luc constructs can measure TGF-β pathway activation by NS3

    • Western blotting for phosphorylated SMAD proteins can assess pathway activation

    • Real-time PCR for fibrogenic genes (collagens, TIMPs) can measure downstream effects

  • Co-localization studies:

    • Immunofluorescence microscopy can visualize NS3 and TGF-β receptor co-localization

    • The effect of inflammatory cytokines (e.g., TNF-α) on this co-localization can be assessed

    • Time-course experiments can track the dynamics of receptor binding and internalization

  • In vivo studies:

    • HCV-infected chimeric mice can be treated with anti-NS3 antibodies targeting predicted receptor binding sites

    • Liver fibrosis can be assessed histologically and through fibrosis marker measurements

    • Correlation between NS3 inhibition and fibrosis reduction can establish causality

Research has shown that NS3 protease can mimic TGF-β2 and directly bind to TGF-β type I receptor (TβRI), thereby enhancing liver fibrosis. This interaction is facilitated by TNF-α, which increases the colocalization of TβRI with NS3 protease on the surface of HCV-infected cells .

How can researchers design assays to evaluate NS3 antibody specificity across HCV genotypes?

Designing assays to evaluate antibody specificity across HCV genotypes requires systematic approaches:

  • Epitope mapping:

    • Express recombinant NS3 proteins from different genotypes (1a, 1b, 2a, 2c, etc.)

    • Perform Western blotting or ELISA to assess antibody reactivity

    • Use peptide arrays or truncation mutants to identify specific binding regions

    • Compare results with sequence alignments to identify conserved vs. variable epitopes

  • Cross-reactivity testing:

    • Develop a panel of NS3 proteins representing major genotypes

    • Use standardized immunoassay conditions to enable direct comparison

    • Calculate relative binding affinities for each genotype

    • Present data as a cross-reactivity matrix showing percent recognition versus genotype 1a (reference)

  • Functional inhibition assays:

    • Test antibody-mediated inhibition of NS3 enzymatic activities across genotypes

    • Compare IC50 values for protease and helicase inhibition

    • Correlate inhibition with binding affinity to identify functionally important epitopes

  • Structural analysis:

    • Map recognized epitopes onto 3D structural models of NS3 from different genotypes

    • Identify conformational differences that might affect antibody binding

    • Design antibodies targeting conserved structural elements for pan-genotypic activity

Available data shows that antibodies like clone 1878 demonstrate specific recognition patterns, reacting with genotypes 1a, 1b, and 2c but not with 2a. This highlights the importance of careful antibody selection for genotype-specific research applications .

What are the critical parameters to optimize when using biotin-labeled NS3 in enzymatic assays?

When using biotin-labeled NS3 in enzymatic assays, several parameters must be carefully optimized:

  • Buffer composition:

    • pH: Optimal range typically 7.0-7.5 for balanced protease and helicase activities

    • Salt concentration: 50-150 mM NaCl for helicase activity; lower concentrations (25-75 mM) for protease activity

    • Divalent cations: 1-5 mM Mg²⁺ required for ATPase and helicase activities

    • Reducing agents: 1-5 mM DTT or 0.5-1 mM TCEP to maintain cysteine residues in reduced state

  • Substrate selection:

    • For helicase assays: RNA or DNA duplexes with 3' overhangs (18-25 bp optimal length)

    • For protease assays: Peptides containing authentic NS3/4A cleavage sites

    • Labeled substrates: Fluorophore/quencher pairs for real-time monitoring

    • Substrate concentration: Below Km for inhibitor studies, near Km for mechanistic studies

  • Enzyme concentration and immobilization:

    • Determine optimal NS3 concentration (typically 10-50 nM for solution assays)

    • For immobilized formats, optimize density on streptavidin surfaces (100-500 ng/well)

    • Ensure proper orientation for accessibility of active sites

    • Include non-biotinylated NS3 controls to assess specific vs. non-specific binding

  • Reaction conditions:

    • Temperature: 25-37°C (lower temperatures reduce activity but increase stability)

    • Time course: Establish linear range of reaction progress

    • ATP concentration: 1-5 mM for optimal helicase activity

    • Co-factors: Include NS4A peptide (or protein) for full protease activity

Control experiments should include:

  • ATP-dependence verification (omitting ATP should abolish helicase activity)

  • Known inhibitor positive controls

  • Heat-inactivated enzyme negative controls

  • Biotin-only controls to rule out effects of the label itself

How can researchers properly design experiments to study NS3-antibody interactions?

Designing robust experiments to study NS3-antibody interactions requires careful consideration of multiple factors:

  • Antibody characterization:

    • Determine antibody isotype, affinity, and epitope specificity

    • Verify recognition of native vs. denatured NS3 forms

    • Establish cross-reactivity profile across HCV genotypes

    • Prepare Fab fragments to eliminate potential Fc-mediated effects

  • Binding assay optimization:

    • ELISA: Optimize coating concentration of biotin-NS3 (typically 0.5-2 μg/ml)

    • Surface Plasmon Resonance: Use controlled immobilization of biotin-NS3 on streptavidin chips

    • Bio-Layer Interferometry: Establish appropriate antibody concentration ranges

    • Include non-specific binding controls and background subtraction

  • Functional inhibition assessment:

    • Establish dose-response relationships for enzyme inhibition

    • Determine mode of inhibition (competitive, non-competitive, uncompetitive)

    • Calculate inhibition constants (Ki) and IC50 values

    • Compare inhibition of different NS3 activities (protease vs. helicase)

  • Cellular studies:

    • Develop intracellular expression systems for antibodies

    • Use appropriate viral replication models (replicon systems or infectious virus)

    • Establish quantitative readouts (HCV RNA levels, viral protein expression)

    • Include proper controls for antibody expression levels and specificity

An example experimental workflow might include:

  • Initial screening of antibody binding by ELISA

  • Kinetic characterization by SPR

  • Epitope mapping using truncated NS3 constructs

  • Functional inhibition studies in cell-free systems

  • Cellular expression and anti-viral activity assessment

This systematic approach enables comprehensive characterization of NS3-antibody interactions and their potential therapeutic applications .

What controls should be included when studying the effect of NS3 on TGF-β signaling?

When investigating the effect of NS3 on TGF-β signaling and its role in liver fibrosis, the following controls are essential:

  • Protein quality controls:

    • Enzymatically active vs. inactive NS3 (use site-directed mutants)

    • Separate NS3 protease and helicase domains to determine domain-specific effects

    • Endotoxin-free preparations to avoid LPS-mediated inflammatory effects

    • Heat-denatured NS3 to control for structure-dependent effects

  • Signaling pathway specificity controls:

    • Recombinant TGF-β2 as positive control at equivalent molar concentrations

    • TGF-β receptor kinase inhibitors to block receptor-mediated signaling

    • SMAD3 phosphorylation inhibitors to block downstream pathway activation

    • Specific antibodies against TGF-β binding sites on NS3

  • Cell-type controls:

    • Hepatic vs. non-hepatic cell lines to establish tissue specificity

    • TGF-β receptor knockout cells to confirm receptor dependence

    • Primary cells vs. cell lines to validate physiological relevance

    • Cells expressing NS3 vs. cells treated with exogenous NS3

  • Experimental condition controls:

    • Time-course experiments (1-48 hours) to distinguish direct vs. indirect effects

    • Dose-response relationships for both NS3 and TGF-β

    • Co-treatment with inflammatory mediators (e.g., TNF-α) vs. NS3 alone

    • Pathway activation in HCV-infected vs. NS3-only expressing cells

Data analysis should include:

  • Normalization to appropriate housekeeping genes or proteins

  • Statistical comparison of NS3 effects relative to TGF-β2 at equivalent concentrations

  • Correlation analyses between NS3 levels and fibrogenic gene expression

  • Multivariate analysis to identify NS3-specific versus general TGF-β-like effects

What are the common challenges in maintaining biotin-labeled NS3 stability and activity?

Maintaining stability and activity of biotin-labeled NS3 presents several challenges with specific solutions:

  • Storage stability issues:

    • Challenge: Loss of enzymatic activity during storage

    • Solution: Store at -80°C in buffer containing 20-50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1 mM DTT, and 10-20% glycerol

    • Validation: Regularly test enzymatic activity using standard helicase or protease assays

    • Expected outcome: >85% activity retention for 6 months under optimal conditions

  • Aggregation problems:

    • Challenge: Protein aggregation leading to activity loss

    • Solution: Include 0.01-0.05% non-ionic detergents (Tween-20, NP-40) and filter through 0.22 μm membrane before use

    • Validation: Check monodispersity by dynamic light scattering or size exclusion chromatography

    • Expected outcome: >90% monomeric protein with proper storage

  • Oxidation sensitivity:

    • Challenge: Oxidation of cysteine residues affecting protein folding and activity

    • Solution: Maintain reducing environment with freshly prepared DTT or TCEP

    • Validation: Measure free thiol content using Ellman's reagent

    • Expected outcome: Preservation of critical thiol groups and associated activity

  • Biotin-streptavidin interaction issues:

    • Challenge: Decreased binding capacity to streptavidin surfaces

    • Solution: Verify biotin accessibility using HABA assay; ensure biotin groups are not masked by protein folding

    • Validation: Compare binding efficiency to fresh preparations

    • Expected outcome: Consistent binding capacity throughout storage period

Activity monitoring should be performed regularly, with acceptance criteria of >70% of initial activity. Implementing these strategies can extend the functional half-life of biotin-labeled NS3 preparations from weeks to months .

How can researchers overcome challenges in studying NS3 inhibitors across different experimental models?

Studying NS3 inhibitors across different experimental models presents several challenges requiring specific approaches:

  • Potency translation across models:

    • Challenge: Discrepancies between biochemical and cellular potency

    • Solution: Establish correction factors based on parallel testing of reference compounds

    • Methodology: Test inhibitors simultaneously in enzyme assays, replicon systems, and infectious virus models

    • Expected outcome: Predictive algorithms relating biochemical IC50 to cellular EC50

  • Genotype-dependent variations:

    • Challenge: Inhibitor efficacy varies across HCV genotypes

    • Solution: Test against a panel of NS3 proteins/replicons representing major genotypes

    • Methodology: Generate comprehensive cross-genotype susceptibility profiles

    • Expected outcome: Identification of pan-genotypic vs. genotype-specific inhibitors

  • Resistance development:

    • Challenge: Emergence of resistance-associated substitutions

    • Solution: Implement resistance selection studies and deep sequencing

    • Methodology: Culture cells with sub-optimal inhibitor concentrations and sequence NS3 region over time

    • Expected outcome: Identification of resistance pathways and cross-resistance patterns

  • Target engagement verification:

    • Challenge: Confirming that cellular effects are NS3-mediated

    • Solution: Use site-directed mutagenesis to create resistance mutations in wild-type NS3

    • Methodology: Compare inhibitor effects on wild-type vs. mutant NS3 in all assay formats

    • Expected outcome: Confirmation of on-target activity through parallel shifts in potency

This systematic approach enables the development of reliable structure-activity relationships and facilitates the design of improved NS3 inhibitors with broader genotype coverage and higher resistance barriers .

What are the emerging applications of biotin-labeled NS3 in understanding HCV-host interactions?

Biotin-labeled NS3 is enabling several emerging applications for studying HCV-host interactions:

  • Proteomic identification of host interaction partners:

    • Pull-down assays with biotin-NS3 coupled to streptavidin beads

    • Mass spectrometry identification of binding partners

    • Validation through reciprocal co-immunoprecipitation

    • Functional classification of interactors to identify key pathways affected by NS3

  • Single-molecule analysis of NS3 helicase mechanisms:

    • Immobilization of biotin-NS3 on streptavidin-coated surfaces

    • Real-time visualization of fluorescently labeled RNA/DNA substrate unwinding

    • Determination of step size, processivity, and kinetic parameters

    • Correlation of structural elements with mechanical function

  • Host immune response modulation:

    • Study of NS3 interactions with pattern recognition receptors

    • Analysis of NS3-mediated interference with innate immune signaling

    • Identification of epitopes recognized by neutralizing antibodies

    • Design of immunogens that elicit cross-genotype neutralizing responses

  • Drug discovery applications:

    • Fragment-based screening using immobilized biotin-NS3

    • Identification of allosteric binding sites through structural studies

    • Development of bifunctional molecules targeting multiple NS3 functions

    • Creation of targeted protein degradation approaches for NS3

Recent studies have revealed that NS3 interacts with over 200 host proteins, including components of RNA processing machinery, innate immunity pathways, and vesicular trafficking networks. These interactions may explain many aspects of HCV pathogenesis beyond viral replication, including immune evasion and oncogenic potential .

How can researchers integrate data from NS3 enzymatic, structural, and cellular studies to develop better antivirals?

Integrated approaches to NS3 research can accelerate antiviral development through the following methodological framework:

  • Structure-function correlation:

    • Align enzymatic activity data with high-resolution structural information

    • Map inhibitor binding sites relative to catalytic residues

    • Identify allosteric networks within NS3 that communicate between domains

    • Develop structure-based pharmacophore models for virtual screening

  • Pharmacological integration:

    • Correlate biochemical potency with physicochemical properties

    • Establish cellular permeability and subcellular localization patterns

    • Determine enzyme occupancy requirements for antiviral activity

    • Assess on-target residence time effects on efficacy

  • Resistance barrier analysis:

    • Map resistance mutations on structural models

    • Assess impact on protein stability and enzymatic function

    • Identify genetic barriers to resistance (number of mutations required)

    • Design inhibitors targeting conserved regions with high functional constraints

  • Combination strategy development:

    • Identify synergistic combinations targeting different NS3 functions

    • Test combinations of NS3 inhibitors with other DAA classes

    • Evaluate resistance profiles of combination approaches

    • Develop mathematical models predicting optimal combination ratios

This integrated approach has already yielded several successful NS3 protease inhibitors in clinical use. Future directions include developing agents with dual helicase-protease inhibitory activity, designing compounds that disrupt NS3-host protein interactions, and creating inhibitors that block NS3's role in immune evasion and fibrosis progression .

Product Science Overview

Introduction to Hepatitis C Virus (HCV)

Hepatitis C Virus (HCV) is a significant global health concern, affecting millions of people worldwide. It is a single-stranded positive-sense RNA virus that primarily targets the liver, leading to chronic hepatitis, cirrhosis, and hepatocellular carcinoma if left untreated . The HCV genome encodes a single polyprotein, which is processed into structural and nonstructural proteins, including the nonstructural protein 3 (NS3) .

NS3 Protein

The NS3 protein of HCV is a multifunctional enzyme with protease and helicase activities. It plays a crucial role in the viral life cycle by processing the viral polyprotein and unwinding RNA structures . The NS3 protein is essential for viral replication and is a target for antiviral drug development .

Recombinant NS3 Protein

Recombinant NS3 protein is produced using genetic engineering techniques, where the NS3 gene is cloned and expressed in a host organism, such as Escherichia coli . The recombinant protein can be purified and used for various research and diagnostic purposes. One such variant is the NS3 protein tagged with biotin, which allows for easy detection and purification using streptavidin-based methods .

Applications of Recombinant NS3 Protein
  1. Research: Recombinant NS3 protein is widely used in research to study the structure and function of the NS3 enzyme. It helps in understanding the mechanisms of viral replication and the development of antiviral drugs .
  2. Diagnostics: The NS3 protein is used in diagnostic assays to detect HCV infection. It serves as an antigen in enzyme-linked immunosorbent assays (ELISA) and other immunoassays .
  3. Vaccine Development: NS3 protein is a potential candidate for HCV vaccine development. DNA vaccines expressing NS3 protein have shown promise in inducing immune responses in preclinical studies .

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.