HCV NS5A

Hepatitis C Virus NS5A Recombinant
Shipped with Ice Packs
In Stock

Description

RNA Replication

NS5A organizes the membranous web (MW) for RNA synthesis by recruiting NS3/4A, NS5B, and host factors:

ProcessMechanismHost Proteins InvolvedReferences
MW formationRemodels ER membranes via PI4KIIIα activationPI4KIIIα, NS4B, NS5B
RNA replicationModulates RNA-binding and polymerase activityNS5B, VAP-A/B, CypA

Key Insight: NS5A inhibitors (e.g., daclatasvir) block MW formation, halting RNA replication .

Virus Assembly

NS5A coordinates Core protein, LDs, and NS5B to facilitate virion packaging:

  • Lipid droplet (LD) interaction: NS5A DI mutants disrupt LD co-localization with Core, impairing assembly .

  • Mitochondrial fragmentation: NS5A induces PI4KIIIα-dependent mitochondrial fission, enhancing apoptosis resistance .

Modulation of Host Cell Pathways

NS5A subverts cellular defenses and stress responses:

Host PathwayNS5A MechanismOutcomeReferences
PKR inhibitionAntagonizes PKR-mediated eIF2α phosphorylationSuppresses IFN-α/β antiviral response
Calcium signalingDisrupts ER Ca²⁺ storage → oxidative stressActivates NF-κB/STAT-3 pathways
AutophagyActivates p62-dependent mitophagyDegrades pro-oxidant proteins (Core, NS5A)

Experimental Evidence:

  • PKR silencing rescues infectious virus production in NS5A DI mutants .

  • Antioxidants (NAC, PDTC) reverse NS5A-induced oxidative stress and transcription factor activation .

  • Mitophagy degrades damaged mitochondria, linked to HCV-associated liver diseases .

Drug Resistance and Therapeutic Targeting

NS5A inhibitors are cornerstone therapies but face resistance challenges:

DrugTargetResistance Mutations (Genotype 1a)EC₅₀References
DaclatasvirDI domainM28T/V, Q30E/R, L31M, Y93H/N50 pM (1a)
VelpatasvirDI/DII domainsA92K, P97S, Y93H9 pM (1b)

Clinical Relevance:

  • NS5A resistance testing (e.g., Labcorp’s GT1a NS5A Profile) identifies mutations at positions 28, 30, 31, 93 .

  • Combination therapies (e.g., NS5A + protease inhibitors) overcome resistance by targeting multiple steps .

Research Advances and Unresolved Questions

  • NS5A dimerization: Surface residues (e.g., F148, W243) mediate self-interaction, critical for replication and CypA binding .

  • Autophagy dynamics: NS5A-induced mitophagy may promote HCC by modulating ROS and cancer stem cell niches .

  • Immune evasion: NS5A inhibits PKR/IRF1 pathways, but its role in innate immune modulation remains under investigation .

Product Specs

Introduction
NS5A is a component of the replication complex that participates in RNA-binding. The hyperphosphorylated form of the NS5A protein inhibits viral replication. NS5A interacts with human VAPB, targeting the viral replication complex to vesicles. Additionally, NS5A downregulates viral IRES translation initiation and mediates interferon resistance by interacting with and inhibiting human EIF2AK2/PKR. NS5a also inhibits apoptosis by interacting with BIN1 and FKBP8.
Description
Recombinant HCV NS5A, produced in E. coli, is a single polypeptide chain consisting of 118 amino acids (aa 2210-2306) with a molecular weight of 13.4 kDa. It is fused to a 21 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
A sterile, colorless solution.
Formulation
The HCV NS5A protein solution (0.25 mg/ml) is supplied in 20mM Tris-HCl buffer (pH 8.0), 0.2M NaCl, 40% glycerol, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), store at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is greater than 90% as determined by SDS-PAGE analysis.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSLKATCTAN HDSPDAELIE ANLLWRQEMG GNITRVESEN KVVILDSFDP LVAEEDEREV SVPAEILRKS RRFARALPVW ARPDYNPPLV ETWKKPDY.

Q&A

What is the structural organization of NS5A and how does it relate to function?

NS5A is organized into three distinct domains (I, II, and III), separated by two repetitive low complexity sequence stretches. The N-terminal Domain I contains a conserved zinc-binding site that functions as a structural metal ion critical for RNA replication . NS5A is peripherally anchored to membranes by an N-terminal amphipathic helix and adopts a dimeric structure that represents a novel class of protein fold . The dimeric orientation creates a large groove between monomers that serves as a putative RNA binding site . This structural arrangement is essential for NS5A's multiple functions in viral replication and assembly.

How does NS5A dimerization correlate with HCV replication?

NS5A dimerization directly correlates with its RNA binding activity and HCV replication. The protein forms dimers that are believed to bind RNA, which is essential for viral replication . Glutaraldehyde cross-linking experiments have demonstrated that RNA binding requires NS5A dimerization . The large groove formed between the two monomers in the dimeric structure provides an optimal RNA interaction surface . Research suggests that NS5A inhibitors may suppress viral replication by preventing dimer/oligomer formation, thereby disrupting RNA binding . This mechanistic understanding has informed the development of potent antiviral compounds targeting the dimerization interface.

What RNA sequences does NS5A preferentially bind and how does this affect viral replication?

NS5A represents a novel structural class of RNA-binding proteins with highest affinity for G/U-rich RNA or G/U-rich elements (GREs) that are five to six nucleotides in length . The optimal RNA-binding region maps to domain I and the first low-complexity sequence of NS5A (domain I-plus) . G/U-rich RNA promotes dimerization of domain I-plus, consistent with the NS5A dimer structure which contains a groove of positive electrostatic potential with residues capable of hydrogen bonding to guanine and uracil bases . One putative viral RNA-binding site for NS5A is the poly(rU) tract in the 3′-UTR of the HCV genome . This specificity suggests NS5A may selectively interact with particular regions of the viral genome during replication.

What are the key phosphorylation states of NS5A and their functional significance?

NS5A exists in two primary phosphorylation states: a basally phosphorylated form and a hyperphosphorylated form . These can be visualized on Western blots as two separate bands, with the upper band representing the hyperphosphorylated form and the lower band representing the hypophosphorylated form . The phosphorylation status significantly impacts viral function, with cell culture-adaptive mutations that reduce NS5A hyperphosphorylation conferring efficient replication of genotype 1 replicons . Conversely, suppression of hyperphosphorylation through kinase inhibitors or mutagenesis allows higher RNA replication in non-culture-adapted replicons, while inhibiting replication in adapted replicons . This indicates a complex regulatory role of NS5A phosphorylation in the viral life cycle.

Which host kinases are responsible for NS5A hyperphosphorylation?

Through high-throughput screening of 404 human protein kinases, CKI-α (casein kinase I-alpha) has been identified as a key NS5A-associated kinase involved in NS5A hyperphosphorylation . CKI-α-dependent hyperphosphorylation influences viral production more profoundly through virion assembly than through viral replication . Mechanistically, this hyperphosphorylation recruits NS5A to low-density membrane structures around lipid droplets and facilitates its interaction with the core protein for new virus particle formation . Proteomic approaches have identified the region within the low-complexity sequence I of NS5A involved in hyperphosphorylation and its regulation of infectious virus production .

How can researchers experimentally manipulate NS5A phosphorylation to study its effects?

Researchers can manipulate NS5A phosphorylation through several experimental approaches:

  • Site-directed mutagenesis of serine residues, particularly in the C-terminal domain III where alanine replacements of serine clusters impair phosphorylation

  • Application of specific kinase inhibitors targeting CKI-α or other kinases involved in NS5A phosphorylation

  • Modulation of host factors like Amphiphysin II (which interacts with NS5A residues 350-356) that regulate NS5A phosphorylation

  • Use of culture-adaptive mutations that naturally reduce hyperphosphorylation

  • Expression of phosphomimetic mutants using aspartic or glutamic acid substitutions at key phosphorylation sites

Each approach provides different insights into how phosphorylation regulates NS5A function in the viral life cycle.

How does NS5A contribute to HCV RNA replication?

NS5A is absolutely required for HCV RNA replication, though its precise mechanistic contribution remains incompletely defined . It colocalizes with viral RNA in stable cell lines expressing the HCV replicon, suggesting direct involvement in replication complexes . The protein's RNA binding activity, particularly its affinity for G/U-rich elements that are abundant in the viral genome, likely plays a critical role . NS5A interacts with numerous host factors essential for replication and may serve as a scaffold protein organizing the viral replication complex. The N-terminal Domain I and its zinc-binding site are particularly important for RNA replication functions .

What is the precise role of NS5A in virion assembly and how does it differ from its replication functions?

NS5A's role in virion assembly is distinct from its replication functions and primarily involves the C-terminal domain III, which is not essential for viral RNA replication but critical for producing infectious virus . Alanine replacements of serine clusters in domain III impair NS5A phosphorylation, leading to decreased NS5A-core protein interaction, perturbed subcellular distribution, and disrupted virion production . NS5A facilitates virion assembly through interactions with host factors like Annexin A2 (via domain 3) and Apolipoprotein E (via residues 205-280) . The protein appears to function as a bridge between replication complexes and assembly sites, potentially delivering newly synthesized viral RNA to core proteins at lipid droplets where assembly occurs.

What methodological approaches can distinguish between NS5A's roles in replication versus assembly?

Researchers can differentiate between NS5A's replication and assembly functions through:

  • Domain-specific mutagenesis (targeting domain I for replication effects versus domain III for assembly effects)

  • Trans-complementation assays where NS5A variants are expressed separately from the viral polyprotein

  • Time-course analyses separating early replication events from later assembly steps

  • Subcellular fractionation to isolate replication complexes versus assembly sites

  • Use of assembly-defective but replication-competent viral constructs

  • Monitoring of both viral RNA levels (replication) and infectious particle production (assembly)

  • Confocal microscopy to visualize NS5A colocalization with either replication markers or assembly markers

These approaches allow researchers to dissect the multifunctional nature of NS5A in the viral life cycle.

What is the interactome of NS5A and how does it influence viral functions?

NS5A interacts with a diverse array of host proteins that contribute to its multifunctional nature. The table below summarizes key interaction partners identified in research:

ProteinNS5A interaction residuesFunctional role
2-5A synthase1-148Inhibition of IFN signaling
ActinMovement of replication complexes
Akt
Amphiphysin II350-356Modulation of NS5A phosphorylation
Annexin A2domain 3Assembly of infectious virus
Apo-AIPromotion of HCV-induced steatosis
Apo-E205-280Assembly of infectious virus
BAX262-277Inhibition of apoptosis
β-Catenin1-144Inhibition of apoptosis
Myc box-dependent-interacting protein 1348-356Inhibition of apoptosis

These interactions collectively enable NS5A to modulate host cell processes to facilitate viral replication, evade immune responses, prevent apoptosis, and promote assembly of infectious particles .

How does the NS5A-Cyclophilin A interaction influence HCV replication?

Cyclophilin A (CypA) is a critical host factor that interacts with NS5A and significantly impacts HCV replication . While the search results don't detail the specific mechanism, experimental approaches such as GST-pulldown assays with recombinant GST-CypA protein are used to study this interaction . CypA is believed to function as a peptidyl-prolyl isomerase that may induce conformational changes in NS5A, potentially affecting its interactions with other viral and host proteins or its RNA-binding capabilities. This interaction represents a potential therapeutic target, as cyclophilin inhibitors have demonstrated antiviral activity against HCV.

What experimental protocols are most effective for identifying novel NS5A-host protein interactions?

For identifying and characterizing NS5A-host protein interactions, researchers can employ several methodological approaches:

  • GST pulldown studies: Incubate glutathione beads in dialysis buffer (50 mM Tris, pH 7.4, 100 mM NaCl, 5 mM MgCl₂, 10% glycerol, 0.5% Nonidet P-40) with BSA, then wash in binding buffer (20 mM Tris, pH 7.9, 0.5 M NaCl, 10% glycerol, 1% Nonidet P-40)

  • Co-immunoprecipitation followed by mass spectrometry for unbiased discovery

  • Yeast two-hybrid screening to identify binary interactions

  • Proximity-based labeling methods (BioID, APEX) to capture transient interactions

  • FRET or BRET assays to study interactions in living cells

  • Surface plasmon resonance or isothermal titration calorimetry to determine binding affinities

  • Mutagenesis approaches to map interaction interfaces with amino acid precision

These approaches provide complementary information about the NS5A interactome.

What are the degradation pathways for NS5A and how do they affect viral persistence?

NS5A is degraded by both major protein degradation pathways of the host cell: the autophagosome (autophagy) pathway and the proteasome pathway . The protein has a relatively short half-life of approximately 2.43 hours in untreated cells, which is much shorter than other viral nonstructural proteins like NS3 (half-life of 24.2 hours) . Inhibiting either degradation pathway increases NS5A stability, with autophagy inhibitors causing a particularly dramatic increase in NS5A half-life . This differential stability may represent a regulatory mechanism allowing the virus to control the relative abundance of its proteins during different stages of infection.

How can researchers experimentally measure NS5A stability and degradation kinetics?

Researchers use cycloheximide (CHX) chase assays to determine NS5A half-life . This methodology involves:

  • Inhibiting protein translation with cycloheximide

  • Collecting cellular samples at multiple time points (e.g., 11 points over 4 hours)

  • Performing Western blot analysis with anti-NS5A antisera

  • Quantifying both hyperphosphorylated (upper band) and hypophosphorylated (lower band) forms

  • Normalizing to housekeeping proteins like β-actin

  • Calculating half-lives through non-linear regression analysis

Additional approaches include pulse-chase labeling with radioactive amino acids, fluorescence-based degradation reporters, and pharmacological inhibition of specific degradation pathways.

What is the relationship between NS5A phosphorylation status and its stability?

While the search results don't explicitly address the relationship between NS5A phosphorylation and stability, the data indicate that NS5A exists in both hyperphosphorylated and hypophosphorylated forms that can be visualized and quantified separately in degradation experiments . Research methodology typically involves quantifying both bands of NS5A in Western blots and calculating their combined half-life . The relationship between phosphorylation and stability represents an important area for further investigation, particularly whether the two phosphorylation states exhibit different degradation kinetics and whether this contributes to the regulation of NS5A function during the viral life cycle.

What are the molecular mechanisms of action for current NS5A inhibitors?

NS5A inhibitors represent a novel class of direct-acting antivirals that target the NS5A protein . While their precise mechanism remains incompletely understood, evidence suggests they may suppress viral RNA replication by preventing NS5A dimer/oligomer formation and disrupting RNA binding . The design of these inhibitors has evolved from promising monomers to highly potent dimeric compounds . Current approved NS5A inhibitors include daclatasvir (Daklinza), elbasvir (in Zepatier), ledipasvir (in Harvoni), ombitasvir (in Viekira Pak/XR and Technivie), pibrentasvir (in Mavyret), and velpatasvir (in Epclusa and Vosevi) .

How do specific mutations in NS5A confer resistance to different classes of NS5A inhibitors?

Specific mutations in the NS5A gene have been implicated in resistance to various HCV antiviral drugs . The Hepatitis C Virus NS5A Drug Resistance assay detects mutations and polymorphisms in HCV genotype 2 associated with resistance to direct-acting antivirals such as pibrentasvir and velpatasvir . Multiple resistance mutations have been characterized, with the levels of increased resistance for each mutation calculated using HCV replicons or reporter constructs . Some mutations confer cross-resistance to multiple NS5A inhibitors . Knowledge of these resistance patterns is crucial for clinical management, as incomplete viral suppression could prevent sustained viral response and promote development of further drug resistance .

What computational and experimental approaches are advancing the design of next-generation NS5A inhibitors?

Researchers are employing sophisticated approaches to design more potent NS5A inhibitors:

  • Quantitative structure-activity relationship (QSAR) modeling to identify structural features enhancing inhibitory activity

  • Monte Carlo optimization techniques to build predictive models of inhibitor efficacy

  • Molecular docking to predict binding affinity within the NS5A protein

  • Molecular dynamics simulations to investigate dynamic interactions over time

  • Molecular mechanics generalized born surface area calculations to estimate binding free energies

  • ADMET (absorption, distribution, metabolism, excretion, toxicity) analyses to assess pharmacokinetic profiles

  • Rational design of dimeric phenylthiazole compounds with enhanced potency

This comprehensive approach provides detailed understanding of potential efficacy, stability, and safety of candidate NS5A inhibitors .

What are the most effective cell culture systems for studying NS5A functions?

While the search results don't explicitly detail all cell culture systems, they mention using stable cell lines expressing the HCV replicon, particularly in Huh-7 cells . Effective experimental systems include:

  • Subgenomic replicon systems in Huh-7 or Huh-7.5 cells

  • Full-length HCV cell culture (HCVcc) systems

  • Trans-complementation systems where NS5A is expressed separately

  • Inducible expression systems to control NS5A levels

  • Reporter-linked systems to monitor viral replication efficiency

  • Primary human hepatocyte cultures for more physiologically relevant conditions

  • 3D organoid models that better recapitulate liver architecture

These systems allow researchers to investigate NS5A functions under different conditions and in various cellular contexts.

How can researchers distinguish between direct and indirect effects of NS5A mutations?

Distinguishing direct from indirect effects of NS5A mutations requires multiple complementary approaches:

  • Site-directed mutagenesis targeting specific functional domains or motifs

  • Revertant analysis to confirm causality between mutations and phenotypes

  • Trans-complementation assays to rescue function with wild-type protein

  • Biochemical assays with purified proteins to assess direct effects on specific functions

  • Time-resolved studies to establish temporal relationships between effects

  • Structural biology approaches to determine how mutations alter protein conformation

  • Systems biology approaches to map broader effects on viral-host interaction networks

By combining these methodologies, researchers can establish causal relationships between NS5A mutations and observed phenotypes.

What are the current challenges and limitations in NS5A research methodologies?

Though not explicitly stated in the search results, several challenges in NS5A research can be inferred:

  • Obtaining sufficient quantities of purified, correctly folded NS5A for structural studies

  • Distinguishing between the multiple functions of this multifunctional protein

  • Capturing transient interactions with host factors or viral components

  • Developing systems that recapitulate the complete viral life cycle

  • Resolving apparently contradictory results regarding phosphorylation effects

  • Determining the precise mechanism of action for NS5A inhibitors

  • Understanding the functional significance of different NS5A conformations

  • Translating in vitro findings to in vivo significance

Addressing these methodological challenges represents an important frontier in advancing our understanding of this critical viral protein.

Product Science Overview

Introduction

Hepatitis C Virus (HCV) is a significant global health concern, affecting over 150 million people worldwide. It is a major cause of chronic liver diseases, including cirrhosis and hepatocellular carcinoma (HCC) . The HCV genome encodes both structural and nonstructural proteins, which play crucial roles in the virus’s life cycle. Among these, the nonstructural protein 5A (NS5A) is particularly important due to its multifunctional nature and its involvement in viral replication and assembly .

Structure and Function of NS5A

NS5A is a phosphoprotein that consists of three distinct domains, each contributing to different aspects of the viral life cycle . The N-terminal amphipathic alpha-helix (amino acids 5–25) anchors NS5A to the endoplasmic reticulum membrane, facilitating the formation of double-membrane vesicles (DMVs) essential for viral replication .

  • Domain I: Contains zinc-binding and RNA-binding motifs crucial for HCV replication .
  • Domain II: Involved in RNA replication and interacts with various host cell factors .
  • Domain III: Essential for virus assembly and release .
Role in Viral Replication and Assembly

NS5A interacts with other viral proteins, such as NS4B and NS5B, and host cell proteins, including cyclophilin A and various kinases, to regulate viral replication and assembly . It plays a critical role in the formation of DMVs, which provide a conducive environment for efficient viral replication .

Recombinant NS5A

Recombinant NS5A proteins are engineered versions of the natural NS5A protein, often used in research to study the protein’s function and to develop antiviral drugs. These recombinant proteins are produced using various expression systems, such as bacterial, yeast, or mammalian cells, to ensure proper folding and functionality.

Applications in Research and Drug Development

Recombinant NS5A proteins are invaluable tools in the study of HCV biology and the development of antiviral therapies. They are used in:

  • Structural Studies: To understand the protein’s three-dimensional structure and its interactions with other molecules .
  • Drug Screening: To identify and test potential inhibitors of NS5A, which can block viral replication .
  • Vaccine Development: To explore the potential of NS5A as a target for vaccine candidates .

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.