HBXIP Human

Hepatitis B Virus x Interacting Protein Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Functional Domains

HBXIP adopts a profilin-like fold with a β-sheet core and α-helical regions, enabling dimerization via anti-parallel β-strands and a leucine zipper motif . This structure facilitates interactions with partners such as survivin, Nrf2, and IRE1α. Key structural features include:

FeatureDescriptionSource
FoldProfilin-like β-sheet core with α-helices
DimerizationForms dimers via extended β-sheets and leucine zipper between α2 helices
Interaction MotifsBinds survivin, Nrf2, XIAP, and IRE1α

HBXIP exists in multiple isoforms, with the shortest (91 amino acids) forming the core structure . Its localization to lysosomal membranes enables participation in the Ragulator complex, which activates mTORC1 signaling .

Oncogenic Functions in Cancer

HBXIP is overexpressed in breast, hepatocellular, and esophageal cancers, where it drives:

FunctionMechanismCancer TypeSource
ProliferationUpregulates PCNA, downregulates p27Breast, Liver
Apoptosis InhibitionBinds pro-caspase-9 via survivin, blocking mitochondrial apoptosisBreast, Liver
Metabolic ReprogrammingSuppresses SCO2/PDHA1 (oxidative phosphorylation) → enhances glycolysisBreast
Lipid SynthesisUpregulates fatty acid synthase (FASN) via miR-183/182/96 clustersBreast

Stress Response Regulation

HBXIP modulates oxidative stress and unfolded protein response (UPR):

  • Nrf2-ARE Pathway: Competes with Nrf2 for Keap1 binding, reducing ROS levels .

  • UPR Inhibition: Stabilizes IRE1α-BiP complex, suppressing proapoptotic UPR signaling in tamoxifen-resistant (TamR) breast cancer .

Expression Patterns in Cancers

HBXIP is highly expressed in aggressive tumors, correlating with poor outcomes:

Cancer TypeExpression LevelCorrelationSource
Breast CancerHigh (87.5% cases)Larger tumors, poor survival (PFS < 80 months)
Hepatocellular CarcinomaHighAdvanced stages, poor prognosis
GliomaHighIncreased immune-cell infiltration, poor OS

Therapeutic Implications

HBXIP’s role in radioresistance and TamR highlights its potential as a therapeutic target:

MechanismTarget AxisClinical ImpactSource
RadioresistanceHBXIP → NF-κB → XIAP ↑Enhances tumor survival post-irradiation
Tamoxifen ResistanceHBXIP → IRE1α inactivationReactivates UPR, restores drug sensitivity

Key Signaling Pathways

HBXIP intersects with multiple pathways to promote malignancy:

PathwayHBXIP’s RoleEffectorSource
mTORC1Activates via Ragulator complex → enhances cell growthLAMTOR1-4
NF-κBUpregulates XIAP → inhibits caspase-3 → radioresistanceXIAP
Nrf2-AREReduces ROS → promotes survival under oxidative stressNrf2
UPR (IRE1α)Stabilizes IRE1α-BiP complex → suppresses apoptosis in TamR cellsIRE1α

Experimental Insights

  • Radioresistance: HBXIP overexpression in MDA-MB-231 cells increased tumor radioresistance in vivo, reversed by XIAP siRNA .

  • Metabolic Reprogramming: HBXIP downregulates SCO2/PDHA1, shifting metabolism to aerobic glycolysis .

Product Specs

Introduction
Hepatitis B virus x interacting protein (HBXIP) interacts with the hepatitis B virus X (HBX) protein's C-terminus, forming a complex. This interaction negatively regulates HBX activity, altering the virus's replication cycle. HBXIP is also involved in cellular processes like bipolar spindle formation, centrosome dynamics regulation, and cytokinesis, potentially through its interaction with Dynein light chain. The protein is highly expressed in skeletal and cardiac muscle, followed by the pancreas, kidney, liver, brain, placenta, and lung. In individuals with chronic HBV infection, HBXIP levels are elevated in both cancerous and non-cancerous liver tissue compared to HBV-free hepatic tissue.
Description
Recombinant human HBXIP, produced in E. coli, is a single polypeptide chain comprising 197 amino acids (aa 1-173) with a molecular weight of 20.7 kDa. HBXIP is tagged at the N-terminus with a 24 amino acid His-tag and purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The HBXIP solution (1mg/ml) is supplied in 20mM Tris-HCl buffer (pH 8.0), 0.1M NaCl, 10% glycerol, and 1mM EDTA.
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 exceeds 90% as determined by SDS-PAGE analysis.
Synonyms
Ragulator complex protein LAMTOR5, Hepatitis B virus X-interacting protein, HBV X-interacting protein, HBX-interacting protein, Late endosomal/lysosomal adaptor and MAPK and MTOR activator 5, LAMTOR5, HBXIP, XIP.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMEPGAG HLDGHRAGSP SLRQALCDGS AVMFSSKERG RCTVINFVPL EAPLRSTPRS RQVTEACGGE GRAVPLGSEP EWSVGGMEAT LEQHLEDTMK NPSIVGVLCT DSQGLNLGCR GTLSDEHAGV ISVLAQQAAK LTSDPTDIPV VCLESDNGNI MIQKHDGITV AVHKMAS.

Q&A

What is HBXIP and where is it located in the human genome?

HBXIP (Hepatitis B virus X-interacting protein) is a protein encoded by the HBXIP gene located on human chromosome 1. The protein was initially identified through its specific interaction with the C-terminus of the hepatitis B virus X protein (HBx) . At the cellular level, HBXIP is predominantly cytoplasmic under normal conditions but can translocate to the nucleus upon DNA damage or other cellular stresses . The protein's functional domains include regions for protein-protein interactions that mediate its diverse cellular functions.

What are the primary molecular functions of HBXIP in normal cells?

HBXIP serves as a multifunctional regulatory protein in normal cells. Its primary molecular functions include:

  • Regulation of viral replication: HBXIP negatively regulates HBx activity, thereby altering the replication cycle of the hepatitis B virus .

  • Cell cycle regulation: HBXIP functions as a mediator of DNA damage response signals, activating G2/M checkpoints to maintain genomic integrity .

  • Apoptosis regulation: HBXIP forms a complex with survivin and inhibits apoptosis via the mitochondrial/cytochrome c pathway by competitively inhibiting the activation of the caspase-9 precursor protein by Apaf1 .

  • Centrosome replication and cell division: Proper HBXIP expression is necessary for normal spindle formation during mitosis; abnormal expression levels can result in single-stage or multi-stage spindles .

For studying these functions, researchers typically employ protein interaction studies, gene knockdown/overexpression experiments, and cell cycle analysis methods.

How does HBXIP interact with other cellular proteins?

HBXIP engages in multiple protein-protein interactions that mediate its diverse cellular functions. Key interactions include:

  • NCOA6 (Nuclear Receptor Coactivator 6): HBXIP has been shown to interact with NCOA6, suggesting a role in transcriptional regulation .

  • Survivin: HBXIP forms a complex with survivin to inhibit apoptosis by preventing the activation of caspase-9 .

  • Suv3p: The HBXIP binding domain is important for mitochondrial import and stability of the Suv3 protein .

  • Cell cycle regulatory proteins: HBXIP interacts with components of the ATM-Chk2 pathway in response to DNA damage .

To investigate these interactions, research methodologies such as co-immunoprecipitation, yeast two-hybrid screening, and proximity ligation assays are commonly employed. RNA immunoprecipitation (RIP) assays have specifically been used to demonstrate HBXIP's interaction with lncRNAs like lncRNA-HEIH and lncRNA-HULC .

How does HBXIP regulate hepatitis B virus replication?

HBXIP regulates HBV replication primarily through its interaction with the HBx protein. As a negative regulator of HBx activity, HBXIP forms a specific complex with the C-terminus of HBx, which subsequently alters the viral replication cycle . HBx is known to promote viral replication by modulating host cell transcription, signal transduction, and DNA repair mechanisms.

To study this regulatory function, researchers can utilize:

  • Viral replication assays with HBV-expressing cell lines

  • Co-immunoprecipitation to assess HBXIP-HBx interactions

  • Site-directed mutagenesis to identify critical binding domains

  • Gene expression analysis before and after HBXIP knockdown/overexpression in HBV-infected cells

The regulation appears to be bidirectional, as HBV infection may also influence HBXIP expression levels, creating a complex feedback loop relevant to viral persistence and pathogenesis .

What is the relationship between HBXIP and long non-coding RNAs in HBV infection?

HBXIP interacts with specific long non-coding RNAs (lncRNAs) during HBV infection, which appears to be significant for disease progression. Research has demonstrated that both lncRNA-HEIH and lncRNA-HULC co-immunoprecipitate with HBXIP, indicating direct physical interactions . These interactions have been documented using RNA Immunoprecipitation (RIP) analyses with antibodies against HBXIP.

Key findings regarding these interactions include:

  • Both lncRNA-HEIH and lncRNA-HULC are upregulated in hepatitis B patients, particularly those with HBV-related hepatocellular carcinoma .

  • HBXIP expression levels are higher in HBV-positive HCC samples compared to HBV-negative HCC samples .

  • The interaction may promote HBV replication and contribute to the development of hepatitis B-related diseases .

To investigate these relationships, researchers should consider:

  • RIP assays to confirm RNA-protein interactions

  • Expression correlation studies between lncRNAs and HBXIP

  • Functional studies with knockdown/overexpression of both HBXIP and the lncRNAs

  • Analysis of downstream pathways affected by these interactions

How does HBXIP regulate the G2/M checkpoint in response to DNA damage?

HBXIP functions as a critical regulator of the G2/M checkpoint following DNA damage. Research shows that HBXIP acts as a mediator protein for DNA damage response signals to activate this checkpoint, thereby maintaining genome integrity and preventing cell death .

The mechanisms involve:

  • Regulation of the ATM-Chk2 signaling pathway: HBXIP knockdown affects the activation of this pathway following DNA damage .

  • Modulation of checkpoint proteins: HBXIP depletion decreases the expression of phospho-Cdc25C, phospho-Cdc2 (Tyr15), and p27, which are essential for G2/M arrest .

  • Nuclear translocation: Upon DNA damage, HBXIP mobilizes from the cytoplasm to the nucleus, suggesting a direct role in the nuclear response to DNA damage .

Experimentally, this can be studied through:

  • Cell cycle analysis using flow cytometry with propidium iodide staining

  • Immunoblotting for phosphorylated checkpoint proteins

  • Immunofluorescence to track HBXIP localization after DNA damage

  • Comet assays to assess DNA damage repair efficiency

What is the impact of HBXIP on DNA damage-induced γH2AX foci formation?

HBXIP significantly influences the formation of γH2AX (phospho-histone H2AX) foci, which are markers of DNA double-strand breaks. Research demonstrates that HBXIP knockdown increases phospho-histone H2AX expression and foci formation after treatment with ionizing radiation (IR) .

This suggests that HBXIP plays a protective role against DNA damage, as its absence leads to:

  • Increased DNA damage accumulation

  • Compromised DNA repair mechanisms

  • Enhanced sensitivity to genotoxic stress

For researchers investigating this phenomenon, methodological approaches include:

  • Immunofluorescence microscopy to quantify γH2AX foci

  • Time-course experiments to assess foci formation and resolution

  • Comet assays to directly measure DNA strand breaks

  • Combined knockdown/overexpression studies with components of DNA repair pathways

The relationship between HBXIP and γH2AX provides important insights into how HBXIP contributes to genome stability and potentially to resistance against DNA-damaging therapeutic agents in cancer cells.

How is HBXIP expression altered across different cancer types?

HBXIP demonstrates consistent upregulation across multiple cancer types compared to corresponding normal tissues, suggesting a common oncogenic role. The expression patterns vary by cancer type and have been documented through extensive tissue analyses.

Cancer TypeHBXIP ExpressionDetection MethodsReference
Breast cancerHighly expressed in MCF-7 and SK-BR3 cells; not detected in epithelial MCF-10A cellsWestern blot, RT-qPCR
Bladder urothelial carcinomaElevated in T24 and PC3 cellsImmunohistochemistry, Western blot
Gastric cancerElevated compared to normal gastric mucosa and adjacent tissueTissue microarray, RT-qPCR
Liver cancer (HCC)Significantly upregulated, particularly in HBV-positive HCCWestern blot, immunohistochemistry
Ovarian cancerHighly expressedImmunohistochemistry

Research methodologies for studying HBXIP expression in cancer include:

  • RT-qPCR for mRNA quantification

  • Western blotting and immunohistochemistry for protein detection

  • Tissue microarrays for high-throughput analysis

  • Correlation analyses with clinical parameters and patient outcomes

The consistent overexpression across diverse cancer types indicates that HBXIP upregulation may be a common mechanism in oncogenesis, making it a potential biomarker and therapeutic target .

What molecular mechanisms underlie HBXIP's promotion of cancer cell proliferation?

HBXIP promotes cancer cell proliferation through multiple molecular mechanisms that collectively enhance cell cycle progression and inhibit apoptosis. These mechanisms include:

  • Modulation of cell cycle regulators: HBXIP upregulates cyclin-D1 and cyclin-E expression while inhibiting p21 and p27 expression, promoting G1/S phase transition in liver and breast cancer cells .

  • Activation of signaling pathways: HBXIP activates the PI3K/Akt pathway in hepatocellular carcinoma cells, increasing cyclin-D1 and phosphorylated protein kinase B while downregulating p53 and p21 .

  • Inhibition of apoptosis: HBXIP forms a complex with survivin to competitively inhibit caspase-9 activation by Apaf1, blocking mitochondria-mediated cell apoptosis .

  • Transcriptional regulation: HBXIP promotes cell proliferation by modulating transcriptional factor Sp1 and HDAC6 in human cancer cells .

  • Angiogenesis promotion: HBXIP enhances angiogenesis in hepatocellular carcinoma, contributing to tumor growth .

To investigate these mechanisms, researchers commonly employ:

  • Proliferation assays (MTT, BrdU incorporation)

  • Cell cycle analysis by flow cytometry

  • Western blotting for pathway components

  • Chromatin immunoprecipitation for transcriptional targets

  • RNA interference to validate specific mechanisms

  • Xenograft models to confirm in vivo relevance

How does HBXIP influence chemosensitivity in cancer cells?

HBXIP significantly impacts cancer cell response to chemotherapy, with evidence suggesting that its downregulation sensitizes cancer cells to chemotherapeutic agents. This relationship has important implications for cancer treatment strategies and overcoming therapy resistance.

Research has demonstrated that:

  • HBXIP knockdown increases cancer cell sensitivity to chemotherapy .

  • This enhanced sensitivity is accompanied by increased apoptosis and cleavage of caspase-3 and caspase-9 .

  • HBXIP's regulatory effect on the G2/M checkpoint may contribute to chemoresistance, as this checkpoint allows cells to repair DNA damage before cell division .

Experimental approaches to study HBXIP's influence on chemosensitivity include:

  • Cell viability assays with dose-response curves to chemotherapeutic agents

  • Combination studies with HBXIP inhibition and chemotherapy

  • Apoptosis detection using Annexin V/PI staining and flow cytometry

  • Western blotting for apoptotic markers

  • Colony formation assays to assess long-term survival after treatment

Understanding HBXIP's role in chemosensitivity may reveal opportunities for combination therapies that target HBXIP alongside conventional chemotherapeutics to enhance treatment efficacy.

What are the most effective approaches for studying HBXIP protein interactions?

Investigating HBXIP protein interactions requires robust methodologies that can capture both stable and transient interactions across different cellular compartments. The most effective approaches include:

  • Co-immunoprecipitation (Co-IP): The gold standard for protein-protein interaction studies, particularly useful for identifying HBXIP binding partners like survivin, HBx, and components of the ATM-Chk2 pathway .

    • Protocol considerations: Use of crosslinking agents, optimization of lysis buffers, and validation with reciprocal Co-IP

  • RNA Immunoprecipitation (RIP): Essential for studying HBXIP interactions with RNA molecules like lncRNA-HEIH and lncRNA-HULC .

    • Implementation: Using Magna RIP RNA-Binding Protein Immunoprecipitation Kit with antibodies against HBXIP

  • Proximity Ligation Assay (PLA): Valuable for visualizing and quantifying protein interactions in situ with high sensitivity.

    • Advantages: Detects endogenous proteins, provides spatial information about interactions

  • FRET/BRET: For studying dynamic interactions and conformational changes in living cells.

    • Applications: Real-time monitoring of HBXIP interactions under various cellular stresses

  • Yeast Two-Hybrid Screening: Useful for identifying novel interaction partners of HBXIP.

    • Follow-up: Validation in mammalian cells with the above methods

  • Mass Spectrometry-Based Interactomics: For unbiased identification of HBXIP interaction networks.

    • Protocol: Tandem affinity purification followed by mass spectrometry analysis

Each method has specific advantages and limitations, and researchers should consider using complementary approaches to comprehensively characterize HBXIP interactions in their specific biological context.

What genetic manipulation techniques are most appropriate for HBXIP functional studies?

Functional characterization of HBXIP requires effective genetic manipulation techniques to modulate its expression and activity. Based on current research practices, the following approaches are most appropriate:

  • RNA Interference (RNAi):

    • siRNA transfection has been successfully used to knockdown HBXIP expression in multiple cancer cell lines

    • shRNA for stable knockdown in long-term experiments and in vivo studies

    • Optimization of transfection conditions is critical for high efficiency and specificity

  • CRISPR-Cas9 Genome Editing:

    • For complete knockout of HBXIP or introduction of specific mutations

    • Can be used to tag endogenous HBXIP with reporters for localization studies

    • Design multiple guide RNAs targeting different exons for validation

  • Overexpression Systems:

    • Plasmid-based expression with inducible promoters for controlled expression

    • Viral vectors (lentivirus, adenovirus) for efficient delivery to difficult-to-transfect cells

    • Fusion constructs with epitope tags or fluorescent proteins for detection and localization studies

  • Domain-Specific Mutations:

    • Structure-function studies using point mutations or domain deletions

    • Particularly useful for examining specific interactions with HBx, survivin, or lncRNAs

  • Rescue Experiments:

    • Re-expression of wild-type or mutant HBXIP in knockdown/knockout backgrounds

    • Critical for confirming specificity of observed phenotypes

When designing these experiments, researchers should consider:

  • Cell type-specific differences in HBXIP expression and function

  • Potential compensatory mechanisms in long-term studies

  • Validation of knockdown/overexpression efficiency at both mRNA and protein levels

  • Phenotypic characterization across multiple cellular processes (proliferation, apoptosis, DNA damage response)

How might targeting HBXIP be leveraged for cancer therapeutics?

Targeting HBXIP presents a promising therapeutic strategy for cancer treatment, particularly given its overexpression in multiple cancer types and its involvement in critical cellular processes. Several approaches could be developed:

  • Direct HBXIP Inhibition:

    • Small molecule inhibitors targeting HBXIP protein-protein interactions

    • Peptide-based inhibitors that disrupt specific HBXIP complexes

    • RNA-based therapeutics (siRNA, antisense oligonucleotides) to reduce HBXIP expression

  • Synthetic Lethality Approaches:

    • Identifying and targeting pathways that become essential in HBXIP-overexpressing cancers

    • Combination with DNA-damaging agents, as HBXIP knockdown increases sensitivity to chemotherapy

  • Targeting Upstream Regulators:

    • Inhibition of transcription factors or signaling pathways that drive HBXIP expression

    • Modulation of lncRNAs that interact with HBXIP

  • Immunotherapeutic Approaches:

    • Development of HBXIP-targeting antibodies or immune cell therapies

    • Exploration of HBXIP as a tumor-associated antigen for cancer vaccines

Research indicates that HBXIP inhibition sensitizes cancer cells to chemotherapy, as evidenced by increased apoptosis and cleavage of caspase-3 and caspase-9 . This suggests that combining HBXIP inhibition with conventional chemotherapeutics could enhance treatment efficacy and potentially overcome resistance mechanisms.

Future therapeutic development should consider:

  • Cancer type-specific functions of HBXIP

  • Potential toxicity in normal cells

  • Delivery methods to ensure target engagement

  • Biomarkers to identify patients most likely to respond to HBXIP-targeted therapies

What are the challenges in developing HBXIP as a biomarker for HBV-related diseases?

Developing HBXIP as a biomarker for HBV-related diseases faces several technical and biological challenges that researchers must address:

  • Expression Heterogeneity:

    • HBXIP expression varies across different stages of HBV infection and HBV-related diseases

    • Requires standardized quantification methods and established clinical thresholds

  • Sample Collection and Processing:

    • Need for minimally invasive sampling methods (beyond liver biopsies)

    • Stability of HBXIP in different sample types and storage conditions

    • Standardization of extraction and detection protocols

  • Analytical Validation:

    • Establishing sensitivity, specificity, reproducibility, and accuracy of HBXIP detection methods

    • Correlation between tissue and circulating HBXIP levels

    • Distinguishing HBV-specific changes from general inflammatory responses

  • Clinical Validation:

    • Large-scale prospective studies linking HBXIP levels to disease progression and outcomes

    • Integration with existing biomarkers (HBsAg, HBV DNA, ALT/AST) for improved predictive value

    • Determining the additive value over current diagnostic approaches

  • Biological Complexity:

    • HBXIP interactions with lncRNAs (lncRNA-HEIH and lncRNA-HULC) add complexity to biomarker development

    • Need to consider both protein levels and functional activity

    • HBV genotype variations may affect HBXIP interactions and expression

Research approaches to address these challenges include:

  • Development of sensitive ELISA or multiplexed protein assays for HBXIP detection in serum

  • Multi-omics approaches combining HBXIP with other molecular markers

  • Longitudinal studies tracking HBXIP expression from chronic HBV infection through disease progression

  • Integration of HBXIP data with clinical parameters for risk stratification models

Overcoming these challenges could establish HBXIP as a valuable biomarker for early detection, prognosis, and therapeutic response prediction in HBV-related diseases.

Product Science Overview

Introduction

Hepatitis B Virus (HBV) is a significant human pathogen that causes liver inflammation, cirrhosis, and hepatocellular carcinoma. One of the critical proteins encoded by HBV is the Hepatitis B Virus X protein (HBx), which plays a crucial role in the virus’s life cycle and its interaction with host cells .

Hepatitis B Virus X Protein (HBx)

HBx is a 154-amino acid protein that facilitates the efficient replication of HBV by stimulating HBV gene expression from the covalently closed circular DNA (cccDNA) template . The exact mechanisms by which HBx interacts with host proteins and facilitates HBV replication are still under investigation. However, it is known that HBx can interact with various host cellular factors to modulate viral and cellular gene expression .

Interaction with Host Proteins

HBx interacts with several host proteins, which can either promote or inhibit HBV replication. One such interaction is with the cellular protein CBFβ, which has been shown to inhibit HBV replication by blocking the formation of the HBx-CUL4-DDB1-SMC complex . This complex is essential for the degradation of the host restriction factor SMC5/6, which suppresses HBV transcription from the cccDNA template .

Another important interaction is with the novel X-associated cellular protein XAP2. XAP2 is a cytoplasmic protein that inhibits the transactivation function of HBx, thereby acting as a negative regulator of HBx . The interaction between HBx and XAP2 requires a small region on HBx containing amino acids 13–26 .

Recombinant HBx Protein

Recombinant HBx protein is produced using recombinant DNA technology, which involves inserting the HBx gene into an expression vector and introducing it into a host cell, such as Escherichia coli or yeast. The host cells then produce the HBx protein, which can be purified and used for various research purposes. Recombinant HBx protein is valuable for studying the interactions between HBx and host proteins, as well as for developing potential therapeutic interventions targeting HBx .

Role in Hepatocellular Carcinoma

HBx is also implicated in the development of hepatocellular carcinoma (HCC). It acts as an indirect transcriptional transactivator, regulating the expression of many viral and cellular genes . HBx has been shown to promote cell proliferation and inhibit apoptosis, contributing to the development and progression of HCC . The exact mechanisms by which HBx contributes to HCC are still being elucidated, but it is clear that its interactions with host proteins play a significant role.

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.