HCV Core NS3

Hepatitis C Virus Nucleocapsid (core) NS3 Recombinant
Shipped with Ice Packs
In Stock

Description

Recombinant HCV protein produced in E.Coli containing the Core (130 a.a.) and NS3 (236 a.a.) excluding NS4 and NS5 domains having a total Mw of 45kDa and purified by proprietary chromatographic techniques.

Product Specs

Introduction
HCV, a member of the Flaviviridae family, is a small (50nm), enveloped virus containing a single-stranded, positive-sense RNA genome. Characterized by a high replication rate, HCV produces roughly one trillion particles daily within an infected individual. Its RNA polymerase lacks proofreading ability, resulting in a high mutation rate, which is thought to contribute to its ability to evade the host's immune response. HCV is classified into six genotypes (1-6), each with multiple subtypes. The distribution and prevalence of these genotypes vary globally. Genotype is a clinically significant factor influencing the potential response to interferon-based therapy and the required treatment duration. Genotypes 1 and 4 exhibit lower responsiveness to interferon-based treatments compared to genotypes 2, 3, 5, and 6.
Description
This product consists of a recombinant HCV protein produced in E. coli. The protein encompasses the Core (130 amino acids) and NS3 (236 amino acids) regions, excluding the NS4 and NS5 domains, resulting in a total molecular weight of 45kDa. The protein has been purified using proprietary chromatographic techniques.
Physical Appearance
The product appears as a clear, colorless solution that has been sterilized through filtration.
Formulation
The product is provided as a sterile-filtered solution containing phosphate-buffered saline (PBS), 1M urea, and 25mM arginine.
Purity
The purity of the product exceeds 95.0%, as determined by SDS-PAGE analysis.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freeze-thaw cycles should be avoided.
Source
Escherichia Coli.

Q&A

What are the structural and functional characteristics of HCV Core and NS3 proteins?

HCV Core is a 21 kDa highly conserved protein that forms the viral capsid subunit. It exhibits multiple roles beyond structural assembly, including interactions with cellular pathways that contribute to pathogenesis .

NS3 is a 67 kDa multifunctional protein with two distinct functional domains: an N-terminal serine protease domain and a C-terminal NTPase/helicase domain. The C-terminal region houses seven canonical SF2 helicase motifs (I, Ia, and II through VI) within domains 1 and 2 . Notable features include the Arg-clamp motif connecting domains IV and V, and the Phe-loop situated between motifs V and VI. The ATP binding site lies between domains 1 and 2, forming two RecA-like domains . The protease activity of NS3 requires NS4A as a cofactor for full enzymatic function .

Both proteins have been implicated in pathogenesis beyond their viral replication functions, particularly in oxidative stress modulation and carcinogenesis pathways.

How do Core and NS3 proteins interact during the HCV lifecycle?

Core and NS3 proteins demonstrate critical genetic and functional interactions during the HCV lifecycle. Research has identified that residues 64-66 of Core's D1 domain form a highly specific interaction with the NS3 helicase domain that is essential for generating infectious HCV particles at a stage downstream of nucleocapsid assembly .

This interaction was revealed through mutational studies where modifications to Core residues 64-66 prevented production of infectious virions despite normal nucleocapsid assembly. Intriguingly, a compensatory mutation (K1302R) within the NS3 helicase domain completely rescued virus production in the context of mutated Core, while the same NS3 mutation abrogated virus production with wild-type Core protein . This indicates a precise structural complementarity between specific residues in these proteins that is essential for viral assembly and maturation.

What cellular pathways are affected by HCV NS3 protein expression?

NS3 protein affects multiple cellular pathways contributing to both viral replication and pathogenesis:

  • EGFR signaling pathway: NS3/4A cleaves T-cell protein tyrosine phosphatase (TC-PTP), a negative regulator of EGFR signaling. This cleavage results in activation of both EGFR and protein kinase B (Akt), which promotes viral replication and resistance to apoptosis .

  • Oxidative stress response: Cells expressing NS3/4A show altered reactive oxygen species (ROS) production and increased resistance to oxidative stress-induced apoptosis .

  • ER stress pathways: NS3/4A expression induces endoplasmic reticulum stress markers GRP78 (HSPA5) and sXBP1 at levels comparable to chemical ER stress inducers like tunicamycin. Interestingly, when NS3/4A-expressing cells are additionally exposed to oxidative stress, this ER stress response is significantly reduced .

  • Neuregulin 1 pathway: NS3 can cleave neuregulin 1 (NRG1), potentially contributing to EGFR pathway upregulation in HCV-infected cells .

What experimental approaches can best elucidate the mechanism of NS3-mediated carcinogenesis?

To investigate NS3's role in hepatocarcinogenesis, researchers should employ a multi-faceted experimental approach:

  • In vitro transformation assays: Express NS3 (or its domains separately) in cell lines like NIH3T3 fibroblasts or primary hepatocytes to monitor transformation-associated characteristics such as:

    • Contact inhibition

    • Doubling time

    • Cloning efficiency

    • Anchorage-independent growth in soft agar

    • Focus formation

  • Proteomic interaction studies: Use co-immunoprecipitation, proximity ligation assays, and mass spectrometry to identify NS3 binding partners within cancer-related pathways.

  • Domain-specific mutagenesis: Create targeted mutations in different NS3 domains to determine which specific regions and activities (protease vs. helicase) contribute to cellular transformation.

  • Signaling pathway analysis: Examine the phosphorylation status of key carcinogenic signaling nodes (MAPK, Akt, STAT) in response to NS3 expression through Western blotting and reverse-phase protein arrays.

  • In vivo models: Develop transgenic mice with hepatocyte-specific NS3 expression to assess long-term carcinogenic potential and compare with Core-expressing models.

  • Clinical correlation studies: Analyze NS3 sequence variations from HCV patients at different disease stages (chronic infection, cirrhosis, HCC) to identify polymorphisms associated with cancer progression, such as the Tyr1082/Gln1112 polymorphism already linked to increased HCC risk .

How can researchers effectively measure the impact of NS3 on oxidative stress and apoptosis resistance?

To comprehensively evaluate NS3's effects on oxidative stress and apoptosis, researchers should implement these methodological approaches:

  • ROS measurement paradigm:

    • Total cellular ROS using fluorescent probes like DCFDA

    • Mitochondrial superoxide using MitoSOX

    • Sequential stress testing (measure baseline NS3 effects, then challenge with oxidative stress inducers like menadione)

    • Include appropriate controls (empty vector transfection, antioxidant treatment with NAC)

  • Apoptosis assessment:

    • Measure caspase-3 activity in NS3-expressing cells before and after oxidative challenge

    • Flow cytometry with Annexin V/PI staining

    • TUNEL assay for DNA fragmentation

    • Western blot analysis of apoptotic markers (cleaved PARP, Bcl-2/Bax ratio)

  • Gene expression analysis:

    • Quantify antioxidant response genes (HO-1, SOD1, SOD2) through qRT-PCR

    • Evaluate ER stress markers (GRP78/HSPA5, sXBP1)

    • Compare expression in sorted cell populations to account for transfection efficiency variability

  • Experimental design considerations:

    • Use multiple cell models (hepatoma cell lines and primary hepatocytes)

    • Control transfection efficiency through co-expression markers like GFP

    • Confirm protein expression through Western blot and immunofluorescence

    • Include pharmacological inhibitors to validate pathway involvement

MethodMeasurementControl ConditionsSignificance
DCFDA fluorescenceTotal cellular ROSEmpty vector, NAC treatmentDetects broad oxidative status
MitoSOXMitochondrial superoxideEmpty vector, NAC treatmentSpecific to mitochondrial ROS
Caspase-3 activity assayApoptosisEmpty vector, NAC reversalQuantifies primary apoptotic effector
qRT-PCRAntioxidant gene expressionEmpty vector, tunicamycinReveals adaptive transcriptional response

What are the methodological considerations when investigating Core-NS3 protein interactions?

When studying the interactions between HCV Core and NS3 proteins, researchers should consider these methodological approaches:

  • Mutagenesis strategy:

    • Create targeted mutations in Core (particularly residues 64-66) and compensatory mutations in NS3 (like K1302R)

    • Design mutations that alter interaction without disrupting protein folding

    • Generate complementary mutations to test genetic interaction hypotheses

  • Viral production assays:

    • Use both intracellular and extracellular infectivity assays to distinguish between assembly and release defects

    • Employ isopycnic gradient analyses to examine nucleocapsid formation and density

    • Quantify viral RNA and protein levels to ensure expression is comparable between constructs

  • Protein-protein interaction detection:

    • Co-immunoprecipitation with antibodies against Core or NS3

    • FRET or BRET assays to detect interactions in living cells

    • In vitro binding assays with purified proteins to determine direct interactions

    • Crosslinking mass spectrometry to identify precise interaction interfaces

  • Structural biology approaches:

    • Cryo-EM of assembled particles with wild-type or mutant proteins

    • Hydrogen-deuterium exchange mass spectrometry to map interaction surfaces

    • Molecular dynamics simulations to predict consequences of mutations

  • Cell biology assays:

    • Co-localization studies using confocal microscopy

    • Live-cell imaging to track dynamic interactions during viral lifecycle

    • Subcellular fractionation to determine compartment-specific interactions

How can researchers investigate the role of NS3 in modulating immune responses?

NS3's involvement in immune modulation requires specialized experimental approaches:

  • Innate immune signaling assessment:

    • Measure activation of pattern recognition receptors (RIG-I, MDA5, TLR3) in presence/absence of NS3

    • Assess IRF3/7 phosphorylation and nuclear translocation

    • Quantify type I interferon production and downstream ISG induction

    • Evaluate NS3's effect on adaptor proteins like MAVS and TRIF

  • Epitope-specific T cell response analysis:

    • Generate T cell clones specific for NS3 epitopes (e.g., region 1248-1261)

    • Measure T cell activation markers, proliferation, and cytokine production

    • Compare responses between NS3 variants from different HCV genotypes

    • Analyze peptide binding to MHC molecules and TCR recognition

  • Genotype-specific immunological differences:

    • Compare immune responses to NS3 from genotype 1b (less immunological) versus 1a and 3a

    • Characterize serological reactivity patterns to different NS3 variants

    • Correlate NS3 secondary structure variations with immune evasion capacity

  • Clinical correlation approaches:

    • Analyze patient samples to correlate NS3 sequence variations with immune markers

    • Compare antibody responses targeting different NS3 domains across disease stages

    • Longitudinal studies tracking NS3 epitope evolution and corresponding T cell responses

What techniques are most effective for expressing and purifying functional NS3 protein for biochemical studies?

For obtaining functional NS3 protein for biochemical studies, researchers should consider these methodological approaches:

  • Expression system selection:

    • Bacterial expression: E. coli BL21(DE3) with pET vectors for high yield

    • Baculovirus/insect cell system: For better folding and post-translational modifications

    • Mammalian cell expression: For studies requiring authentic processing and modification

  • Construct design considerations:

    • Express full-length NS3 with NS4A cofactor (or NS4A peptide) for protease activity

    • Use domain-specific constructs for focused studies on protease or helicase

    • Include purification tags (His6, GST, MBP) positioned to minimize functional interference

    • Consider codon optimization for the chosen expression system

  • Purification strategy:

    • Implement multi-step purification:

      • Initial affinity chromatography (Ni-NTA, glutathione)

      • Ion exchange chromatography

      • Size exclusion chromatography for final polishing

    • Optimize buffer conditions (pH, salt, glycerol) to maintain stability

    • Consider adding reducing agents to prevent oxidation of catalytic cysteines

  • Activity verification assays:

    • Protease activity: Fluorogenic peptide substrates containing NS3/4A cleavage sites

    • Helicase activity: DNA or RNA unwinding assays using fluorescently labeled substrates

    • ATPase activity: Colorimetric assays measuring phosphate release

  • Storage considerations:

    • Determine optimal buffer composition for long-term stability

    • Test cryopreservation conditions (glycerol percentage, flash freezing)

    • Validate activity retention after storage

What are the experimental challenges in studying the synergistic effects of Core and NS3 on host cell metabolism?

Investigating the combined effects of Core and NS3 on host metabolism presents several methodological challenges that require careful experimental design:

  • Expression system considerations:

    • Develop systems for controlled co-expression at physiologically relevant ratios

    • Options include:

      • Dual promoter plasmids

      • Bicistronic constructs

      • Inducible expression systems with orthogonal inducers

    • Confirm protein expression levels through Western blotting with protein-specific antibodies

  • Metabolic analysis approaches:

    • Implement targeted and untargeted metabolomics to detect alterations in:

      • Lipid metabolism (Core primarily affects this)

      • Glucose utilization

      • Redox balance (both proteins modulate this)

    • Measure metabolic flux using isotope-labeled substrates

    • Assess mitochondrial function through respiration analysis

  • Confounding factors to control:

    • Transfection efficiency variability (60% ± 4.8 for cell lines, 9.5% ± 3.2 to 16% ± 4.2 for primary cells)

    • Cytotoxic effects from protein expression

    • Endogenous oxidative stress from transfection procedures

    • Cell density and growth phase effects on metabolism

  • Cellular model selection trade-offs:

    • Hepatoma cell lines (Huh-7): Higher transfection efficiency but altered baseline metabolism

    • Primary hepatocytes: More physiologically relevant but lower transfection efficiency and shorter viability

    • Transgenic systems: Stable expression but potential adaptation effects

  • Mechanistic dissection approach:

    • Use domain-specific mutants to attribute observed effects to specific protein functions

    • Implement pharmacological inhibitors to validate pathway involvement

    • Conduct gene silencing (siRNA) of key metabolic regulators to determine essentiality

How can researchers effectively compare the oncogenic potential of NS3 versus other HCV proteins?

To systematically compare the oncogenic potential of NS3 versus other HCV proteins (particularly Core and NS5A), researchers should implement these methodological approaches:

  • Standardized transformation models:

    • Express individual HCV proteins in multiple cell models:

      • Immortalized hepatocytes (maintains hepatocyte characteristics)

      • NIH3T3 fibroblasts (standard transformation assay)

      • Primary hepatocytes (most physiologically relevant)

    • Ensure equivalent expression levels through calibrated expression systems

    • Include combined expression conditions to detect cooperative effects

  • Comprehensive transformation assessment:

    • Evaluate multiple hallmarks of transformation systematically:

      Transformation CharacteristicMeasurement MethodExpected NS3 Effect
      Contact inhibitionFocus formation assayReduced with N-terminal protease domain
      Growth rateDoubling time measurementIncreased
      Anchorage independenceSoft agar colony formationEnhanced
      Tumorigenic potentialXenograft models in micePositive with NS3 5' segment
      Genomic instabilityKaryotype analysisIncreased
      Invasion capacityTranswell migration assayEnhanced
  • Molecular mechanism comparison:

    • Perform comparative transcriptomics (RNA-seq) and proteomics analyses

    • Identify protein-specific and shared dysregulated pathways

    • Validate key pathways through targeted inhibition studies

    • Compare effects on established cancer-related pathways:

      • EGFR/Akt signaling (NS3 cleaves TC-PTP)

      • Wnt/β-catenin pathway

      • p53 pathway

      • Inflammatory signaling

  • Domain-specific oncogenic contribution:

    • Create chimeric proteins swapping domains between viral proteins

    • Test domain-specific mutants (e.g., protease-dead NS3)

    • Identify minimal regions sufficient for transformation

What are the key contradictions in the literature regarding NS3's role in oxidative stress and how to resolve them?

The literature presents several apparent contradictions regarding NS3's effects on oxidative stress that require methodological approaches to resolve:

  • Contradiction in ROS production:

    • Some studies report NS3 as a direct inducer of oxidative stress

    • Other research (including search result ) shows NS3/4A expression attenuates menadione-induced ROS production and protects against oxidative stress-induced apoptosis

    Resolution methodologies:

    • Distinguish between acute vs. chronic effects through time-course experiments

    • Separate direct NS3 effects from adaptive cellular responses

    • Standardize ROS measurement methods (total cellular vs. mitochondrial-specific)

    • Control for expression levels across studies

  • Cell type-specific effects:

    • Differential responses observed between cell lines and primary hepatocytes

    • Variable effects across different hepatocyte-derived cell lines

    Resolution methodologies:

    • Conduct parallel experiments in multiple cell types

    • Characterize baseline antioxidant capacity of different cellular models

    • Account for differences in NS3 expression efficiency (9.5% ± 3.2 to 16% ± 4.2 in primary cells vs. 60% ± 4.8 in cell lines)

  • Contextual dependency of NS3 effects:

    • NS3 appears pro-oxidant in some contexts but protective in others (particularly when cells face secondary stress)

    • ER stress induction by NS3/4A is reduced when cells are additionally treated with oxidative stressors

    Resolution methodology: Design experiments with sequential stress application:

    • Baseline measurements

    • NS3 expression effects alone

    • Combined NS3 + external stressor effects

    • Recovery phase assessment

  • Experimental table to resolve contradictions:

    Experimental ConditionMeasurementCell ModelsExpected OutcomeInterpretation
    NS3 expression aloneTotal ROS & mitochondrial ROSMultiple hepatocyte modelsMild increaseDirect effect
    NS3 + menadione challengeTotal ROS & mitochondrial ROSMultiple hepatocyte modelsLower than control + menadioneAdaptive response
    NS3 + NAC treatmentApoptosis markersHuh-7 cellsRestoration of apoptotic sensitivityROS dependency of protection
    Time-course experimentHO-1, SOD1, SOD2 expressionPrimary hepatocytesEarly induction, later normalizationAdaptation mechanism

How can researchers overcome the challenges of studying Core-NS3 interactions in the context of complete viral replication?

Studying Core-NS3 interactions within complete viral replication presents several methodological challenges that can be addressed through these approaches:

  • System selection considerations:

    • JFH1-based cell culture systems enable complete viral lifecycle studies

    • Consider adapted strains like JFH-AM1 that enhance infectious virus production

    • Sub-genomic replicons allow focused interaction studies without full virion production

  • Interaction disruption strategies:

    • Targeted mutagenesis of interaction interfaces (e.g., Core residues 64-66)

    • Compensatory mutation approaches (e.g., NS3 K1302R mutation)

    • Trans-complementation assays to rescue defective phenotypes

  • Temporal dynamics assessment:

    • Time-course experiments capturing different phases of viral lifecycle

    • Synchronized infection methods to improve resolution of interaction timing

    • Inducible expression systems to manipulate protein availability at specific timepoints

  • Spatial interaction mapping:

    • Super-resolution microscopy to visualize co-localization patterns

    • Proximity ligation assays to detect in situ protein interactions

    • Subcellular fractionation to identify compartment-specific interactions

  • Functional validation approaches:

    • Measure multiple viral lifecycle stages separately:

      • RNA replication (qRT-PCR)

      • Protein expression (Western blot)

      • Nucleocapsid assembly (density gradient analysis)

      • Virion production (infectivity assays)

    • Design competition assays with peptides mimicking interaction domains

  • Technical obstacles and solutions:

    ChallengeMethodological Solution
    Low transfection efficiency in primary cellsUse lentiviral delivery systems
    Cytotoxicity of viral proteinsImplement inducible expression systems
    Distinguishing direct vs. indirect interactionsUse purified proteins in in vitro binding assays
    Stability of assembled complexesImplement crosslinking approaches before analysis
    Off-target effects of mutationsCreate multiple mutation sets targeting same interface

What emerging technologies could advance our understanding of dynamic Core-NS3 interactions during viral assembly?

Several cutting-edge technologies offer promising approaches to elucidate the dynamic interactions between Core and NS3 during viral assembly:

  • Advanced imaging technologies:

    • Live-cell super-resolution microscopy to visualize interactions in real-time

    • Lattice light-sheet microscopy for extended 3D imaging with reduced phototoxicity

    • Correlative light and electron microscopy (CLEM) to connect protein interactions with ultrastructural changes

  • Protein engineering approaches:

    • Split fluorescent protein complementation optimized for viral proteins

    • FRET/FLIM sensors designed for Core-NS3 interaction dynamics

    • Conditionally stable protein domains to control protein availability

  • Structural biology innovations:

    • Cryo-electron tomography of intact infected cells to visualize assembly sites

    • Time-resolved cryo-EM to capture assembly intermediates

    • Integrative structural biology combining multiple data types:

      • X-ray crystallography

      • NMR

      • Mass spectrometry

      • Computational modeling

  • Single-molecule techniques:

    • Single-molecule FRET to measure conformational changes during interactions

    • Optical tweezers to assess binding forces between proteins

    • Single-molecule tracking in living cells to follow interaction dynamics

  • Systems biology approaches:

    • Multi-omics integration (transcriptomics, proteomics, metabolomics) to build comprehensive interaction networks

    • Mathematical modeling of assembly dynamics incorporating Core-NS3 interactions

    • Machine learning analysis of high-dimensional interaction data

How might understanding Core-NS3 interactions contribute to novel antiviral therapeutic approaches?

Understanding Core-NS3 interactions offers several promising avenues for therapeutic development:

  • Direct interaction inhibitor development:

    • Design peptide mimetics targeting the Core residues 64-66 interface with NS3 helicase

    • Develop small molecule inhibitors that bind the interaction pocket

    • Create stapled peptides that offer improved stability and cellular uptake

    • Screen natural product libraries for compounds that disrupt the interaction

  • Allosteric modulator strategies:

    • Target sites that indirectly affect the conformation of interaction interfaces

    • Develop compounds that lock NS3 in conformations incompatible with Core binding

    • Design modulators that affect the dynamics rather than static binding

  • Mutation-specific approaches:

    • Develop genotype-specific therapies targeting NS3 polymorphisms associated with increased pathogenicity

    • Create inhibitors effective against NS3 resistance mutations

    • Design combination therapies targeting multiple viral protein interactions

  • Host-targeting therapeutic strategies:

    • Identify and target host factors required for Core-NS3 interaction

    • Modulate cellular pathways affected by the interaction (oxidative stress, ER stress)

    • Develop interventions that enhance beneficial adaptations while blocking pathogenic ones

  • Therapeutic development workflow:

    Development StageMethodological ApproachExpected Outcome
    Target validationCRISPR screening for host factorsIdentification of essential interaction cofactors
    Compound screeningHigh-throughput fluorescence-based interaction assaysLead compound identification
    Mechanism confirmationResistance mutation analysisBinding site confirmation
    Efficacy testingInfectious virus production assaysQuantification of antiviral potency
    Combination assessmentSynergy testing with existing antiviralsOptimal therapeutic combinations

Product Science Overview

Introduction

Hepatitis C virus (HCV) is a significant global health concern, affecting millions of people worldwide. It is a member of the Hepacivirus genus within the Flaviviridae family. The HCV genome is a single-stranded positive-sense RNA of approximately 9.6 kb in length, encoding a single polyprotein that is processed into structural and non-structural proteins .

Structure and Function of HCV Proteins

The HCV polyprotein is co- and post-translationally processed by cellular and viral proteases to yield 11 viral proteins. These include three structural proteins (core, E1, and E2), a small polypeptide named p7, and six non-structural (NS) proteins (NS2, NS3, NS4A, NS4B, NS5A, and NS5B) .

  • Core Protein: The core protein forms the nucleocapsid, which encapsulates the viral RNA. It plays a crucial role in the assembly and release of the virus.
  • NS3 Protein: The NS3 protein is a multifunctional enzyme with serine protease and helicase activities. It is responsible for cleaving the viral polyprotein to release non-structural proteins and plays a role in subverting the host immune response .
Recombinant NS3 Protein

Recombinant NS3 protein is produced using various expression systems, such as E. coli, to study its structure and function. The NS3 protein is crucial for viral replication and pathogenesis, making it a target for antiviral drug development .

Immunogenicity and Vaccine Development

The core and NS3 proteins are major immunogenic proteins in HCV infection. They elicit strong humoral and cellular immune responses, making them potential candidates for vaccine development . Various strategies, such as incorporating multiple viral proteins and molecular tags, have been employed to optimize the efficacy of HCV DNA vaccines .

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.