HBV-X, His Tag

Hepatitis B Virus X Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

HBV-X Protein

HBV-X is a 154-amino-acid multifunctional protein encoded by the adr4 strain of hepatitis B virus (HBV). It plays a central role in viral transcription, immune evasion, and hepatocarcinogenesis . Key features include:

  • Transcriptional activation: Enhances viral and cellular gene expression via interaction with host transcription factors (e.g., NF-κB, AP-1) .

  • Immune modulation: Suppresses innate immune responses by degrading MAVS, a critical component of the RIG-I-MAVS antiviral pathway .

  • Oncogenic potential: Promotes chromatin remodeling and DNA damage responses, contributing to liver cancer development .

His Tag

The His Tag is a polyhistidine sequence (typically 6×His) appended to recombinant proteins for affinity chromatography. Key properties include:

  • Purification efficiency: Binds to immobilized metal affinity chromatography (IMAC) resins via imidazole side chains .

  • Flexibility: Compatible with N- or C-terminal fusion, allowing minimal interference with protein function .

PropertyHBV-X (adr4)His Tag (6×His)
Size/Sequence154 aa (2–154 aa)6×His (6 residues)
FunctionViral transcription, immune evasionPurification via IMAC
Expression HostE. coli E. coli

His Tag Integration

The His Tag is typically appended to the N- or C-terminus of HBV-X without disrupting its native function. For example:

  • Expression in E. coli: Recombinant HBV-X-His Tag is expressed with >90% purity and low endotoxin levels, enabling high-yield purification .

  • Functional validation: Studies confirm that the His Tag does not interfere with HBV-X’s ability to interact with host proteins like DDB1 or MAVS .

Viral Transcription Activation

HBV-X counteracts host restriction factors to enhance cccDNA transcription:

  • Smc5/6 degradation: Recruits DDB1 to target the Smc5/6 complex for proteasomal degradation, lifting transcriptional repression .

  • Chromatin remodeling: Acetylates histones via p300/CBP, promoting viral promoter accessibility .

  • Transcriptional coactivation: Enhances androgen receptor (AR)-mediated transcription in a DHT-concentration-dependent manner, explaining male predominance in HBV-associated HCC .

Immune Evasion

HBV-X disrupts innate immunity:

  • MAVS degradation: Promotes ubiquitination and proteasomal degradation of MAVS, inhibiting RIG-I-MAVS signaling and IFN-β production .

  • Apoptosis modulation: Binds CFLAR to regulate DISC formation and mitochondrial membrane potential loss, though effects vary by cell type .

Oncogenic Interactions

HBV-X interacts with host proteins to drive hepatocarcinogenesis:

  • AR activation: Synergizes with AR to enhance colony formation in hepatocytes, particularly under high androgen levels .

  • DNA damage response: Disrupts SMC5/6-mediated DNA repair, contributing to genomic instability .

Host Protein Interactions

HBV-X binds diverse host factors, as identified in GST pull-down and mass spectrometry studies:

Host ProteinInteraction DomainFunctional RoleSource
DDB1C-terminal (aa 138–154)Targets Smc5/6 for degradation
CFLARN-terminal (aa 1–50)Modulates DISC and apoptosis
MAVSTransactivation domain (aa 61–137)Promotes ubiquitination and degradation
ARC-terminal (aa 138–154)Enhances androgen-dependent transcription
Bcl-xLBH3-like motif (aa 120–123)Inhibits apoptosis

Zinc-Binding Motif

  • CCCH motif: Residues C61, C69, C137, and H139 coordinate zinc, critical for HBV-X’s interaction with DDB1 and Smc5/6 degradation .

  • Functional validation: Mutations in C61/C69/C137 abrogate HBV-X’s ability to degrade Smc5/6 and activate transcription .

Transactivation Domain

  • Proteomics screening: GST pull-down assays identified 189 host proteins interacting with HBV-X’s transactivation domain, including Pin1 (peptidyl-prolyl cis-trans isomerase) .

  • Functional impact: Pin1 modulates HBV replication, highlighting its therapeutic potential .

Targeting HBV-X

  • Smc5/6 stabilization: Restoring Smc5/6 levels via HBV-X inhibitors could suppress cccDNA transcription .

  • AR pathway modulation: Inhibiting HBV-X-AR synergy may reduce male-biased HCC risk .

  • Zinc chelation: Disrupting the CCCH motif with zinc-binding agents could impair HBV-X function .

Diagnostic Utility

The His Tag enables:

  • High-purity HBV-X production: Facilitates structural studies (e.g., crystallography of HBV-X-Bcl-xL complexes) .

  • Antibody development: Recombinant HBV-X-His Tag serves as an antigen for serological assays .

Product Specs

Introduction

The Hepatitis B virus X protein (HBx), with a molecular weight of 17 kDa, functions as a transcriptional coactivator and plays a crucial role in regulating genes associated with inflammation and cell survival. It exerts its regulatory effects on various transcription factors, including NF-kappaB, and is heavily implicated in the development of liver cancer (hepatocarcinogenesis). Notably, HBx facilitates the binding of cAMP response element binding protein (CREB) to its corresponding response element. Furthermore, HBx stabilizes the cellular coactivator ASC-2 via direct protein-protein interaction, thereby influencing the regulation of genes actively transcribed in liver cancer cells. It also activates both the JNK and MAPK signaling pathways, contributing to the mobilization of cytosolic Ca2+. The interaction between HBx and the general transcription factor TFIIB represents another mechanism underlying its transcriptional transactivation activity. Importantly, HBx has been shown to downregulate the expression of PTEN, a known tumor suppressor gene and a negative regulator of the phosphatidylinositol 3'-kinase/AKT pathway. The development of hepatocellular carcinoma (HCC) is often linked to hepatitis B virus (HBV) infection, and HBx, in particular, plays a pivotal role in HBV-related HCC. The persistent presence of HBx is critical to the pathogenesis of early-stage HCC, and its expression in the liver during chronic HBV infection may serve as a valuable prognostic marker for HCC development.

Description

Recombinant HBV-X, produced in E. coli, is a single polypeptide chain comprising 165 amino acids (residues 2-154) with a molecular weight of 17.8 kDa. It is fused to a 12 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.

Purity

Exceeds 90%.

Formulation

The protein is filtered through a 0.4 μm filter and subsequently lyophilized from a solution containing 0.5 mg/ml protein in 50 mM acetate buffer (pH 4) and 5% trehalose.

Solubility
To prepare a working stock solution of approximately 0.5 mg/ml, it is recommended to reconstitute the lyophilized protein by adding 0.1 M acetate buffer (pH 4) and ensure complete dissolution. For conversion to a higher pH value, it is advisable to dilute the solution extensively with the appropriate buffer to a concentration of 10 μg/ml. It's important to note that the solubility of HBV X antigen is limited at higher concentrations. Before using this non-sterile product in cell culture, it is essential to filter sterilize your culture media or working solutions containing it.
Stability
For long-term storage, the lyophilized protein should be kept at -20°C. After reconstitution, it is recommended to aliquot the product to minimize repeated freeze-thaw cycles. The reconstituted protein remains stable at 4°C for a limited period; no significant changes have been observed after two weeks of storage at 4°C.
Source
Escherichia Coli.
Amino Acid Sequence

MRGSHHHHHH GSAARVCCQL DPARDVLCLR PVGAESRGRP VSGPFGTLPS PSSSAVPADH GAHLSLRGLP VCAFSSAGPC ALRFTSARRM ETTVNAHQVL PKVLHKRTLG LSAMSTTDLE AYFKDCLFKD WEELGEEIRL KVFVLGGCRH KLVCSPAPCN FFTSA.

Q&A

What is HBV-X protein and what are its primary functions in viral pathogenesis?

HBV-X protein (HBx) is a multifunctional viral protein that plays critical roles in promoting HBV infection and persistence. As a viral transcriptional activator, HBx binds to a conserved ATTGG site within the HBV enhancer II/core promoter (EII/Cp) region and recruits host transcription factors including FOXO3α and PGC1α . This binding activity results in enhanced viral gene expression and genome replication .

HBx primarily localizes to the nucleus of infected cells and has been found to have a relatively short half-life . The protein serves as a key mediator in silencing host antiviral defenses while simultaneously promoting viral transcription . One of its most significant functions is the use of host E3 ubiquitin ligase DDB1 to target the SMC5-SMC6 complex for proteasomal degradation, which prevents this host complex from binding to viral episomal DNA and inhibiting transcription .

Additionally, HBx moderately stimulates transcription of various viral and cellular transcription elements, particularly those containing DNA binding sites for NF-kappa-B, AP-1, AP-2, c-EBP, ATF/CREB, or the calcium-activated factor NF-AT .

How can researchers verify the expression and purification of HBV-X with His tag?

Several methodological approaches can be employed to verify the expression and purification of HBV-X with His tag:

  • Western Blot Analysis: Using monoclonal antibodies specifically developed against HBx epitopes. Research has shown that antibodies targeting the HBx₂₇₋₅₀ region demonstrate superior sensitivity in Western blot applications .

  • ELISA Verification: Enzyme-linked immunosorbent assays using anti-HBx monoclonal antibodies can confirm the presence and concentration of the recombinant protein .

  • Confocal Microscopy: Imaging techniques using fluorescently labeled antibodies can visualize the subcellular localization of HBx-His tag proteins and verify their expression patterns .

  • Binding Assays: Verifying that the recombinant HBx-His protein binds to known interaction partners such as DDB1 can confirm functional integrity .

  • Mass Spectrometry: For definitive identification and characterization of the purified protein and potential post-translational modifications.

How does HBV-X interact with host factors to promote viral persistence?

HBV-X protein establishes complex interactions with host factors to promote viral persistence through multiple mechanisms:

  • HMGA1 Positive Feedback Loop: HBx upregulates the expression of high mobility group AT-hook 1 (HMGA1) by interacting with SP1 transcription factor to activate the HMGA1 promoter . Notably, HMGA1 itself acts as a positive regulator of HBV transcription, creating a positive feedback loop that enhances viral replication .

  • Smc5/6 Complex Degradation: HBx recruits the host E3 ubiquitin ligase DDB1 to target the structural maintenance of chromosomes 5/6 complex (Smc5/6) for proteasomal degradation . Under normal conditions, this host complex would bind to viral episomal DNA and inhibit transcription; HBx circumvents this restriction mechanism .

  • HMGB1 Counteraction: HBx directly interacts with high mobility group box 1 (HMGB1) protein and prevents its binding to cccDNA without affecting steady-state levels of HMGB1 . This interaction prevents HMGB1-mediated epigenetic silencing of the HBV cccDNA minichromosome .

  • Histone Modification Regulation: HBx is essential for maintaining cccDNA in a transcriptionally active state characterized by active histone post-translational modification markers . Without HBx, the cccDNA minichromosome shifts to a heterochromatic state with repressive histone modifications .

These interactions collectively establish HBx as a master regulator that disables host restriction mechanisms while simultaneously activating viral gene expression.

What methods can be used to study the role of zinc binding in HBV-X function?

Zinc binding is critical for HBV-X protein function . Researchers can employ several methodologies to investigate this aspect:

  • Site-Directed Mutagenesis: Creating specific mutations in potential zinc-binding residues within the HBx protein sequence, particularly targeting conserved cysteine and histidine residues that typically coordinate zinc ions.

  • Metal Chelation Assays: Using zinc-specific chelators to sequester zinc and observe the effects on HBx function in various experimental systems.

  • Spectroscopic Analysis: Employing techniques such as circular dichroism or nuclear magnetic resonance to detect conformational changes in HBx structure upon zinc binding or removal.

  • Functional Rescue Experiments: Testing whether exogenous zinc supplementation can restore the activity of HBx proteins compromised by mutation or chelation.

  • Zinc-Binding Affinity Measurements: Using isothermal titration calorimetry or fluorescence spectroscopy to quantify the binding affinity of HBx for zinc ions.

  • Structural Studies: X-ray crystallography or cryo-electron microscopy of HBx in complex with zinc to determine precise binding sites and structural consequences.

What approaches can be used to inhibit HBV-X function as a therapeutic strategy?

Given HBx's critical role in HBV persistence, several research approaches aim to inhibit its function:

  • RNA Interference: Small interfering RNAs (siRNAs) targeting HBx mRNA have been demonstrated to reduce HBx protein levels and facilitate HBV clearance . In mouse models of HBV persistence, targeting endogenous HMGA1 through RNA interference facilitated HBV clearance .

  • Disruption of Protein-Protein Interactions: Developing small molecules or peptides that interfere with HBx interactions with critical host factors such as DDB1, HMGA1, or HMGB1 .

  • Zinc Chelation Strategies: Since zinc binding is essential for HBx function, specific chelation approaches that selectively target HBx-bound zinc could be effective .

  • Targeting the HMGA1-HBV Positive Feedback Loop: The feedback mechanism between HBx and HMGA1 represents a potential therapeutic target, as disrupting this loop could reduce both HBx function and HBV replication .

  • Inhibition of HBx-Mediated Transcriptional Activation: Compounds that prevent HBx from binding to the ATTGG site within the viral enhancer II/core promoter could inhibit HBx-mediated transcriptional activation .

How can researchers effectively detect HBV-X protein in infected cells?

Detection of HBV-X protein in infected cells requires specialized approaches due to its low abundance and short half-life:

  • Monoclonal Antibody Selection: Using highly specific monoclonal antibodies that recognize epitopes within the HBx₂₇₋₅₀ region has proven effective . These antibodies should be validated for specificity against the particular HBV genotype under study, as antibody recognition can vary between genotypes .

  • Confocal Microscopy Protocols: For optimal visualization, fixation with 4% paraformaldehyde followed by permeabilization with 0.1% Triton X-100 provides good results for immunofluorescence detection of HBx .

  • Signal Amplification Techniques: Methods such as tyramide signal amplification can enhance detection sensitivity for low-abundance HBx.

  • Nuclear Fractionation: Since HBx predominantly localizes to the nucleus, nuclear fractionation prior to Western blot analysis can concentrate the protein and improve detection .

  • Time-Course Analysis: Due to the short half-life of HBx, time-course experiments should be carefully designed to capture optimal expression windows post-infection or transfection .

What cell culture systems and viral constructs are most suitable for studying HBV-X function?

Several experimental systems have proven valuable for HBV-X research:

  • Primary Human Hepatocytes (PHH): Representing the most physiologically relevant system for studying HBV infection and HBx function, though challenging to maintain in culture .

  • HepG2/NTCP Cells: Human hepatoma cells engineered to express the sodium taurocholate cotransporting polypeptide (NTCP), which serves as the HBV receptor .

  • Inducible Reporter Cell Lines: Established systems modeling infection with wildtype and HBx-null HBV that secrete HA-tagged HBeAg as a semi-quantitative marker for cccDNA transcription .

  • Viral Constructs:

    • prcccDNA: Recombinant cccDNA plasmids with or without functional HBx (prcccDNA/X⁻)

    • p1.3HBV: HBV replicon plasmid containing a 1.3-copy over-length HBV genome

    • Cre Recombinase Expression Systems: Used in conjunction with prcccDNA to generate functional cccDNA in transfected cells

How can researchers assess the impact of HBV-X on viral transcription and replication?

Multiple methodological approaches can quantify HBx's effect on viral processes:

  • Dual Luciferase Reporter Assays: To measure HBx-mediated activation of viral promoters and enhancers .

  • Chromatin Immunoprecipitation-qPCR (ChIP-qPCR): For analyzing HBx recruitment to viral promoters and associated changes in histone modifications on the cccDNA minichromosome .

  • Southern Blot Analysis: To detect and quantify viral DNA replicative intermediates and assess the impact of HBx on viral replication .

  • Northern Blot or RT-qPCR: For measuring viral RNA transcripts, particularly the 3.5 kb pregenomic RNA .

  • Western Blot Analysis: To determine viral protein expression levels .

  • HBeAg ELISA: Using HBeAg secretion as a surrogate marker for cccDNA transcriptional activity in culture supernatants .

How does HBV-X interact with the cccDNA minichromosome to regulate transcription?

HBV-X regulates cccDNA transcription through complex epigenetic mechanisms:

  • Direct Binding and Recruitment: HBx binds to a conserved ATTGG site within the enhancer II/core promoter (EII/Cp) region of cccDNA . This binding facilitates the recruitment of transcription factors FOXO3α and PGC1α to activate viral gene expression .

  • Epigenetic Modulation: HBx is essential for maintaining cccDNA in a transcriptionally active euchromatic state . ChIP-qPCR studies reveal that HBx promotes active histone post-translational modifications on cccDNA-associated histones .

  • Counteracting Host Restriction Factors: HBx prevents the binding of HMGB1 (high mobility group box 1) to cccDNA, thereby counteracting HMGB1's function as an epigenetic silencer of viral transcription . Without HBx, HMGB1 associates with cccDNA and promotes a heterochromatic state with repressive histone modifications .

  • Smc5/6 Complex Degradation: HBx targets the host Smc5/6 complex for proteasomal degradation through recruitment of the E3 ubiquitin ligase DDB1 . The Smc5/6 complex would otherwise bind to viral episomal DNA and prevent transcription .

What is the significance of the HBV-X and HMGA1 positive feedback loop in viral persistence?

The HBx-HMGA1 positive feedback loop represents a sophisticated viral strategy for maintaining persistent infection:

  • Bidirectional Enhancement: HBx upregulates HMGA1 expression by interacting with SP1 transcription factor to activate the HMGA1 promoter . Conversely, HMGA1 binds to a conserved ATTGG site within the viral enhancer II/core promoter and acts as a positive regulator of HBV transcription .

  • Clinical Correlation: Chronic hepatitis B patients in the immune tolerant phase display both higher intrahepatic HMGA1 protein levels and higher serum HBV markers compared to patients in the inactive carrier phase, supporting the significance of this loop in clinical settings .

  • Therapeutic Implications: This reciprocal regulation suggests that disrupting either component of the loop could potentially break the cycle of viral persistence . Mouse model studies have shown that targeting endogenous HMGA1 through RNA interference facilitated HBV clearance .

  • Molecular Mechanism: HMGA1 recruits transcription factors FOXO3α and PGC1α to the viral promoter, enhancing viral gene expression and genome replication . This mechanism is similar to how HBx itself activates viral transcription, creating redundancy that ensures continued viral expression .

What are the major technical challenges in working with recombinant HBV-X protein?

Researchers face several challenges when working with recombinant HBV-X:

  • Protein Stability: The short half-life of HBx makes it challenging to work with as a purified protein. Storage conditions and buffer compositions must be carefully optimized.

  • Solubility Issues: HBx can form aggregates, affecting functional studies. Addition of appropriate detergents or stabilizing agents may be necessary.

  • Functional Confirmation: Ensuring that recombinant HBx-His tag maintains its native functionality requires careful validation, such as testing its ability to bind known interaction partners like DDB1 .

  • Antibody Specificity: The development of highly specific antibodies against HBx has been historically challenging, requiring careful validation of antibody specificity and sensitivity .

  • Genotype Specificity: HBx sequences vary between HBV genotypes, and antibodies may recognize some genotypes better than others . Researchers should confirm that their detection methods are appropriate for the specific HBx variant under study.

How can researchers overcome limitations in detecting low-abundance HBV-X in infected cells?

Detection of low-abundance HBx requires specialized approaches:

  • Antibody Selection: Use of monoclonal antibodies targeting the HBx₂₇₋₅₀ region has shown superior sensitivity . These should be validated for specificity against the particular HBV genotype under study.

  • Signal Amplification: Employing tyramide signal amplification or other signal enhancement techniques for immunofluorescence and Western blot applications.

  • Subcellular Fractionation: Since HBx predominantly localizes to the nucleus, nuclear extraction can concentrate the protein and improve detection sensitivity .

  • Optimized Timing: Due to HBx's short half-life, careful timing of experiments post-infection or transfection is critical to capture peak expression .

  • Proteasome Inhibition: Brief treatment with proteasome inhibitors may increase HBx levels for detection purposes, though this approach should be used cautiously as it may affect other cellular processes.

  • Advanced Microscopy: Super-resolution microscopy techniques can improve visualization of low-abundance nuclear proteins like HBx.

What controls should be included when studying HBV-X interactions with host factors?

Rigorous controls are essential when investigating HBx interactions:

  • HBx-Null Mutants: Viral constructs with HBx ORF obliterated (e.g., by mutating CAA to TAA at the 8th amino acid) serve as critical negative controls .

  • Domain Mutants: HBx constructs with mutations in specific functional domains can help determine which regions are essential for particular interactions.

  • Competitive Binding Assays: Using purified recombinant proteins to compete with cellular interactions can confirm specificity.

  • Antibody Controls: When performing co-immunoprecipitation, appropriate isotype controls and pre-immune sera should be included.

  • Reciprocal Co-IP: Confirming interactions by precipitating with antibodies against both HBx and the suspected interacting partner increases confidence in results.

  • Expression Level Controls: Ensuring comparable expression levels of HBx across experimental conditions, as overexpression can lead to non-physiological interactions.

  • Functional Validation: Demonstrating that disrupting a specific interaction alters HBx function (e.g., its ability to activate transcription) provides functional relevance to the interaction.

What are promising therapeutic approaches targeting HBV-X currently under investigation?

Several therapeutic strategies targeting HBx are being investigated:

  • siRNA Approaches: Small interfering RNAs targeting HBx mRNA have shown promise in reducing HBx protein levels and facilitating HBV clearance in model systems .

  • Disruption of HBx-Host Protein Interactions: Development of small molecules that prevent HBx from interacting with critical host factors such as DDB1, HMGA1, or HMGB1 .

  • HMGA1-HBx Feedback Loop Targeting: Interventions that disrupt the positive feedback loop between HBx and HMGA1 could potentially break the cycle of viral persistence .

  • Zinc-Binding Inhibitors: Given the importance of zinc binding for HBx function , compounds that interfere with this process could be effective.

  • Epigenetic Modulation: Strategies to promote heterochromatic silencing of the cccDNA minichromosome, mimicking the natural restriction observed in the absence of HBx .

  • Combination Approaches: Targeting HBx in combination with current nucleos(t)ide analogues may provide synergistic effects for achieving viral clearance rather than just viral suppression.

How might structural studies of HBV-X advance therapeutic development?

Detailed structural studies of HBx could significantly advance therapeutic strategies:

  • Structure-Based Drug Design: Determination of the three-dimensional structure of HBx, particularly in complex with key host factors like DDB1, would enable rational design of inhibitors that disrupt these interactions.

  • Identification of Critical Domains: Structural analysis could reveal critical functional domains within HBx that could be targeted with higher specificity.

  • Zinc-Binding Pocket Characterization: Detailed understanding of the zinc coordination site could lead to the development of specific inhibitors that interfere with this essential function.

  • Conformational Dynamics: Studies of HBx conformational changes upon binding to different partners could reveal allosteric sites suitable for therapeutic targeting.

  • Crystallization Approaches: Development of strategies to overcome the challenges in crystallizing HBx, possibly through the use of stabilizing mutations or co-crystallization with binding partners.

What insights might be gained from studying HBV-X across different viral genotypes?

Comparative studies of HBx across HBV genotypes could yield valuable insights:

  • Genotype-Specific Functions: Different HBV genotypes are associated with varying clinical outcomes, and differences in HBx sequence and function may contribute to these variations .

  • Antibody Recognition: Studies have already shown that antibodies may recognize HBx from some genotypes better than others , suggesting structural or antigenic differences.

  • Therapeutic Implications: Genotype-specific differences in HBx function could influence the effectiveness of HBx-targeting therapies across different patient populations.

  • Host Factor Interactions: The strength or specificity of interactions with host factors like HMGA1, DDB1, or HMGB1 might vary between HBx proteins from different genotypes .

  • Evolutionary Insights: Comparing HBx sequences and functions across genotypes could provide insights into the evolutionary pressures shaping this viral protein and its adaptations to host defenses.

Product Science Overview

Introduction

Hepatitis B virus (HBV) is a significant global health concern, affecting approximately 240 million people worldwide. Chronic HBV infection can lead to severe liver diseases, including cirrhosis and hepatocellular carcinoma. The HBV X protein (HBx) is a multifunctional regulatory protein that plays a crucial role in the viral life cycle and pathogenesis. Recombinant HBx proteins, especially those tagged with histidine (His tag), are widely used in research to study the protein’s structure, function, and interactions.

HBx Protein

The HBx protein is a small, non-structural protein encoded by the HBV genome. It is involved in various cellular processes, including transcriptional regulation, signal transduction, and apoptosis. HBx is essential for viral replication and has been implicated in the development of HBV-related liver diseases. Due to its critical role, HBx is a target for therapeutic interventions and vaccine development .

Recombinant HBx Protein

Recombinant HBx proteins are produced using genetic engineering techniques. The gene encoding HBx is cloned into an expression vector, which is then introduced into a host organism, such as Escherichia coli. The host organism expresses the HBx protein, which can be purified and used for various research applications. The His tag is a short sequence of histidine residues added to the protein’s N- or C-terminus to facilitate purification using affinity chromatography .

Applications of Recombinant HBx Protein

Recombinant HBx proteins are used in a wide range of research applications, including:

  • Structural Studies: Understanding the three-dimensional structure of HBx to elucidate its functional mechanisms.
  • Functional Assays: Investigating the role of HBx in viral replication, transcriptional regulation, and interaction with host cellular proteins.
  • Vaccine Development: Exploring the potential of HBx as an immunogen in therapeutic vaccines for chronic HBV infection .
  • Drug Screening: Identifying small molecules or peptides that can inhibit HBx function and potentially serve as antiviral agents.

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 2025 TheBiotek. All Rights Reserved.