HPSE WB

Recombinant Human Heparanase-1 WB Control
Shipped with Ice Packs
In Stock

Description

Composition and Production of HPSE WB

HPSE WB is produced in Chinese Hamster Ovary (CHO) cells and purified via orthogonal chromatography. Key specifications include:

PropertyDetails
Molecular Weight50 kDa (active subunit) + 8 kDa subunit (heterodimer)
Concentration1 μg/ml in LDS-PAGE buffer
Storage Conditions–20°C; avoid freeze-thaw cycles
ApplicationsPositive control for monoclonal/polyclonal anti-HPA1 antibodies

The enzyme is synthesized as a 65 kDa latent precursor that undergoes proteolytic cleavage to form the active 50/8 kDa heterodimer .

Functional Role of HPSE in Disease Pathogenesis

HPSE WB is widely used to study HPSE’s involvement in pathological processes:

Cancer Metastasis and Stemness

  • EMT Regulation: HPSE overexpression in prostate cancer cells (e.g., DU145) reduces E-cadherin and increases vimentin/α-SMA, driving epithelial-mesenchymal transition (EMT) .

  • Tumor Microenvironment: HPSE remodels extracellular matrices, releasing growth factors (e.g., FGF-2, TGF-β) that promote angiogenesis and metastasis .

Renal Fibrosis

  • Chronic Kidney Injury: HPSE inhibition (e.g., Roneparstat) reduces EMT markers (α-SMA, collagen-I) and macrophage polarization in ischemia/reperfusion models, mitigating fibrosis .

Viral Pathogenesis

  • HSV-1 Infection: HPSE enhances NF-κB nuclear translocation, amplifying pro-inflammatory cytokines (IL-1β, TNF-α) while suppressing interferon responses .

Western Blot Validation

HPSE WB serves as a critical control for:

  • Detecting endogenous HPSE in tumor tissues .

  • Validating HPSE knockdown/overexpression in cell lines (e.g., PC3, DU145) .

Therapeutic Development

  • Inhibitor Screening: Cyclophellitol-derived inhibitors (e.g., compound 3) show nanomolar efficacy against HPSE, reducing metastasis in murine models .

  • Antibody Specificity: Polyclonal antibodies (e.g., DF12411) target HPSE in WB/IHC, confirming its upregulation in colorectal cancer immune microenvironments .

Biomarker Potential

  • Prognostic Value: Elevated HPSE correlates with poor survival in pancreatic, prostate, and colorectal cancers .

Product Specs

Introduction

Heparanase is an enzyme, specifically an endo-β-D-glucuronidase, that breaks down heparan sulfate chains found on heparan sulfate proteoglycans (HSPGs) within the extracellular matrix (ECM). This degradation process is crucial for ECM breakdown, facilitating the movement and penetration of tumor cells and inflammatory leukocytes (1,2,3). The breakdown of HSPGs by heparanase releases growth factors and cytokines, which in turn promote cell growth and directed cell movement (4,5). Structurally, heparanase is a two-part molecule (heterodimer) composed of a 50 kDa subunit containing the active site and an 8 kDa subunit. Initially produced in a latent 65 kDa precursor form, it undergoes proteolytic processing to become active (1,6). Heparanase is highly present in myeloid leukocytes like neutrophils, platelets, and the human placenta. Studies have shown elevated levels of human heparanase in various primary tumor types, often correlating with increased tumor invasion, blood vessel formation, and unfortunately, poorer survival rates (7,8).

Description

Recombinant Heparanase protein HPA1 is generated in Chinese hamster ovary (CHO) cells and undergoes purification using multiple distinct chromatography steps.

Formulation

Concentration: 1 microgram per milliliter
Content: 100 nanograms
Buffer: LDS-PAGE buffer (140 mM Tris buffer pH 8.5, 10% Glycerol, 2% LDS, 0.015% EDTA, 1.88% (v/v) of 1% Serva Blue G250 and 0.625% (v/v) of 1% Phenol red).

Applications

Serves as a positive control in western blot analysis.

Preparation Protocol

Utilize 20 microliters of recombinant human heparanase 1 (HPA1) per lane to serve as a control when using either monoclonal anti-HPA1 clone HP3/17 antibodies (Catalog Number: Ins-AB-04001) or polyclonal rabbit anti-HPA1 antibody (Catalog Number: Ins-AB-04002).

Storage Procedures

Store at -20 degrees Celsius. Repeated freezing and thawing cycles should be avoided.

Q&A

What is the optimal sample preparation protocol for HPSE Western blot analysis?

For effective HPSE detection via Western blot, samples should be prepared using a lysis buffer containing 1% Triton X-100, 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1 mM EDTA, and protease inhibitor cocktail. HPSE is particularly sensitive to degradation, so working at 4°C throughout the extraction process is essential. For tissue samples, mechanical homogenization in cold lysis buffer followed by 30-minute incubation on ice and centrifugation at 14,000g for 15 minutes yields optimal results. Protein concentration should be determined using Bradford or BCA assay, with 20-50 μg total protein typically loaded per lane depending on expression levels and antibody sensitivity .

Which antibodies are most reliable for HPSE detection in Western blot?

Several antibodies have demonstrated specificity and sensitivity for HPSE detection in Western blot applications. Primary antibodies against both the 50 kDa active form and the 65 kDa latent form should be considered depending on your research question. When studying BRAF V600E-mutant colorectal cancer samples, antibodies that can detect both forms simultaneously are particularly valuable given the significant expression differences observed in this cancer subtype . It's recommended to validate antibody specificity using positive controls (cells with known HPSE overexpression) and negative controls (HPSE-silenced cells) before proceeding with experimental samples.

What are the most common troubleshooting issues when performing HPSE Western blot?

IssuePotential CauseSolution
Weak or absent bandsInsufficient protein loadingIncrease protein concentration to 40-50 μg
HPSE degradationAdd additional protease inhibitors; maintain cold chain
Inefficient transferOptimize transfer conditions for large proteins (65 kDa)
Multiple non-specific bandsAntibody cross-reactivityIncrease blocking time; try different blocking agent (5% BSA)
Sample degradationPrepare fresh samples; add more protease inhibitors
Inconsistent loadingUneven protein measurementNormalize with structural proteins (β-actin, GAPDH)

Particularly for HPSE detection, inefficient transfer is a common issue due to its relatively large molecular weight. Extended transfer times (90-120 minutes) at lower voltage or overnight transfer at 4°C may improve results for the 65 kDa latent form .

How does HPSE expression differ between BRAF V600E-mutant and wild-type colorectal cancer?

HPSE is significantly overexpressed in BRAF V600E-mutant colorectal cancer compared to wild-type BRAF tumors. Western blot analysis consistently reveals higher protein levels in the mutant group, correlating with transcriptomic data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases . This differential expression has significant clinical implications as high HPSE expression is independently associated with inferior survival specifically in the BRAF V600E-mutant cohort. Research using clinical samples from 172 patients demonstrated that HPSE expression could serve as an independent prognostic biomarker in this aggressive CRC subtype .

What molecular mechanisms link HPSE expression to cell cycle regulation in cancer cells?

HPSE silencing experiments reveal a direct link between HPSE expression and cell cycle regulation in BRAF V600E-mutant colorectal cancer. Western blot analysis following HPSE knockdown shows significant changes in key cell cycle regulators:

  • Decreased phosphorylated Rb protein levels

  • Reduced Cyclin E2 expression

  • Increased p27Kip1 expression

  • Decreased AKT phosphorylation

These molecular changes correspond with G0/G1 phase cell cycle arrest, as confirmed by flow cytometry analysis. Mechanistically, HPSE silencing impedes tumor proliferation by downregulating Cyclin E2 expression via the AKT/p27Kip1 pathway . Rescue experiments demonstrate that overexpression of CCNE2 (encoding Cyclin E2) in HPSE-silenced cells restores cell proliferation ability, confirming the essential role of Cyclin E2 in HPSE-mediated cell cycle progression .

How can Western blot be used to evaluate HPSE inhibitor efficacy in experimental models?

Western blot analysis provides a valuable tool for assessing HPSE inhibitor efficacy beyond enzymatic activity assays. When evaluating small-molecule inhibitors such as isoindole-5-carboxylic acid derivatives or benzoxazole compounds, Western blot can determine:

  • Effects on HPSE protein levels (potential transcriptional/translational regulation)

  • Impact on downstream signaling pathways (AKT/p27Kip1/Cyclin E2 axis)

  • Changes in ECM component expression following inhibitor treatment

For comprehensive evaluation, combine Western blot analysis with functional assays including invasion assays, proliferation measurements, and in vivo xenograft models. This multi-faceted approach has proven effective in characterizing compounds like 4-Bn-RK-682, which demonstrates selective HPSE inhibition with anti-metastatic properties in HT1080 cell lines .

What are the key principles for designing rigorous HPSE expression studies?

Proper experimental design for HPSE expression studies requires attention to replication at the appropriate biological scale. A good experimental design must have at least two critical attributes:

  • Replication at the appropriate biological scale (individual, population, species)

  • Factors of interest must be free from confounding variables

For HPSE Western blot studies specifically, this means:

  • Use biological replicates (different patients/mice/independent cell populations) rather than just technical replicates

  • Include appropriate controls for antibody specificity

  • Control for potential confounding factors like tissue heterogeneity, cancer subtype, and treatment history

  • Perform power analysis to determine adequate sample sizes

  • Design experiments to test causality rather than just correlation

How should researchers design HPSE knockdown validation experiments?

Robust validation of HPSE knockdown efficiency requires careful experimental design and appropriate controls. For Western blot validation:

Experimental GroupControl GroupValidation MetricsSecondary Validation
shRNA/siRNA HPSEScrambled sequence≥70% protein reductionqRT-PCR for mRNA levels
CRISPR-Cas9 HPSE KOWild-type cellsComplete protein absenceEnzymatic activity assay
Inducible knockdownNon-induced cellsDose-dependent reductionImmunofluorescence

Include time-course analysis to determine the optimal timepoint for functional studies post-knockdown. For BRAF V600E-mutant colorectal cancer cell lines like HT29 and RKO, Western blot analysis typically shows maximal HPSE reduction 48-72 hours post-transfection . Always validate knockdown efficiency in each experimental replicate rather than assuming consistent efficiency across experiments.

What considerations are important when using HPSE Western blot for biomarker validation?

When evaluating HPSE as a potential prognostic biomarker, as suggested by its role in BRAF V600E-mutant colorectal cancer , consider these critical experimental design elements:

  • Sample size: Conduct power analysis to determine minimum sample numbers needed for statistical significance

  • Patient stratification: Control for confounding variables (age, stage, treatment history)

  • Sample collection: Standardize collection protocols to minimize pre-analytical variables

  • Multiple methodologies: Validate Western blot findings with orthogonal techniques (IHC, ELISA)

  • Quantification: Use digital image analysis software with appropriate normalization controls

  • Statistical approach: Apply multivariate analysis to assess independent prognostic value

The study examining 172 patients with BRAF V600E-mutant CRC demonstrated that robust experimental design and adequate sample size enabled identification of HPSE as an independent prognostic factor through Kaplan-Meier analysis and Cox regression models .

What is the optimal protocol for detecting both latent and active forms of HPSE?

HPSE exists in both latent (65 kDa) and proteolytically processed active (50 kDa) forms, making detection of both forms critical for comprehensive analysis. The optimal Western blot protocol includes:

  • Gel selection: 8-10% polyacrylamide gels provide optimal separation of both forms

  • Transfer conditions: Wet transfer at 90V for 2 hours or 30V overnight at 4°C

  • Blocking: 5% BSA in TBST for 1-2 hours at room temperature

  • Primary antibody: Use antibodies recognizing epitopes present in both forms or use two separate antibodies

  • Detection: Enhanced chemiluminescence with extended exposure times

  • Controls: Include recombinant latent and active HPSE as positive controls

When studying BRAF V600E-mutant colorectal cancer, the active form typically shows stronger association with aggressive phenotypes and poorer prognosis .

How can researchers simultaneously detect HPSE and associated signaling molecules?

For comprehensive pathway analysis, simultaneous detection of HPSE and its associated signaling molecules (pAKT, p27Kip1, Cyclin E2) provides valuable mechanistic insights. A multiplexed approach requires:

  • Careful primary antibody selection: Choose antibodies from different host species

  • Sequential probing: Start with the least abundant protein (often phosphorylated proteins)

  • Membrane stripping: Use gentle stripping buffers between probing cycles

  • Alternative approach: Run multiple identical gels in parallel when protein sizes are similar

  • Fluorescent detection: Consider fluorescent secondary antibodies for simultaneous detection

When studying the AKT/p27Kip1/Cyclin E2 axis in relation to HPSE expression, this approach enables direct correlation between HPSE levels and activation state of downstream signaling molecules within the same samples .

What considerations are important when comparing HPSE Western blot data across different experimental models?

Comparing HPSE expression across different experimental models (cell lines, patient samples, xenografts) requires careful standardization and consideration of model-specific variables:

  • Loading controls: Use multiple housekeeping proteins (β-actin, GAPDH, tubulin) and total protein staining

  • Normalization method: Apply consistent quantification approaches across all models

  • Sample preparation: Adjust extraction protocols for different sample types while maintaining comparable conditions

  • Antibody validation: Verify antibody performance in each model system independently

  • Inter-experimental calibration: Include common reference samples across experiments

  • Biological context: Consider differences in tumor microenvironment between in vitro and in vivo models

Research comparing HPSE expression in cell models and xenografts demonstrates that while trends may be consistent, absolute expression levels can vary significantly between systems , highlighting the importance of these considerations.

How can Western blot analysis contribute to HPSE inhibitor development?

Western blot analysis provides critical insights beyond enzymatic assays in the development pipeline for small-molecule HPSE inhibitors:

  • Target engagement confirmation: Verify that inhibitors affect not just enzymatic activity but potentially protein stability or expression

  • Mechanism elucidation: Determine whether inhibitors affect HPSE-dependent signaling pathways

  • Selectivity assessment: Evaluate effects on related family members or off-target proteins

  • Pharmacodynamic markers: Identify downstream proteins that can serve as biomarkers of inhibitor activity

  • Resistance mechanisms: Investigate compensatory protein expression changes following inhibitor treatment

This approach has proven valuable in characterizing compounds like 4-Bn-RK-682 and isoindole-5-carboxylic acid derivatives as selective HPSE inhibitors with potential therapeutic applications .

What are the experimental considerations when using Western blot to evaluate HPSE inhibitor effects in different disease models?

Disease ModelSpecial ConsiderationsRecommended ControlsKey Readouts
CancerTumor heterogeneityVehicle-treated matched samplespAKT, Cyclin E2, EMT markers
InflammationTissue-specific extractionDisease-matched untreated samplesInflammatory cytokines, NF-κB
DiabetesGlycosylation effectsMatched non-diabetic samplesInsulin signaling proteins
Viral infectionViral protein interferenceUninfected cells with inhibitorViral load markers

When designing these experiments, ensure adequate biological replication at the appropriate scale and control for potential confounding factors . For cancer models specifically, consider the differential response observed in BRAF-mutant versus wild-type backgrounds .

How might single-cell analysis techniques complement traditional Western blot for HPSE research?

While Western blot provides valuable population-level data on HPSE expression, emerging single-cell technologies offer complementary insights:

  • Cell-to-cell variability: Identify subpopulations with differential HPSE expression that may be masked in bulk analysis

  • Spatial context: Correlate HPSE expression with location within tumor microenvironment

  • Co-expression patterns: Identify cells co-expressing HPSE with specific markers of interest

  • Temporal dynamics: Track HPSE expression changes in individual cells over time

  • Rare cell populations: Detect HPSE-expressing cells that constitute a small fraction of the population

For BRAF V600E-mutant colorectal cancer research, integrating single-cell analysis with traditional Western blot could reveal whether the poor prognosis associated with high HPSE expression stems from uniform elevation or from critical subpopulations of high-expressing cells .

What are the emerging applications of phospho-specific Western blot analysis in HPSE research?

Given the demonstrated role of HPSE in modulating signaling pathways like AKT in BRAF V600E-mutant colorectal cancer , phospho-specific Western blot analysis represents a valuable approach for deeper mechanistic studies:

  • Direct HPSE phosphorylation: Investigate potential regulatory phosphorylation sites on HPSE itself

  • Signaling cascade mapping: Systematically analyze effects of HPSE modulation on phosphorylation states across multiple pathways

  • Kinase inhibitor combinations: Assess synergistic effects between HPSE inhibition and targeted kinase inhibitors

  • Resistance mechanisms: Identify phosphorylation-based compensatory pathways that emerge following HPSE inhibition

  • Biomarker development: Develop phospho-protein signatures as predictive biomarkers for HPSE-targeted therapies

This approach could be particularly valuable in understanding why HPSE silencing affects the AKT/p27Kip1/Cyclin E2 axis and identifying additional targetable nodes in this pathway .

Product Science Overview

Structure and Mechanism

Heparanase is an endo-β-D-glucuronidase that cleaves heparan sulfate (HS) side chains of HSPGs, resulting in shorter oligosaccharide chains . The enzyme is synthesized as a latent 65 kDa precursor, which is proteolytically processed into its active form, consisting of a 50 kDa subunit harboring the active site and an 8 kDa subunit . This heterodimeric structure is essential for its enzymatic activity.

Biological Functions

Heparanase is involved in various physiological and pathological processes, including:

  • ECM Degradation: Facilitates the migration and extravasation of tumor cells and inflammatory leukocytes by degrading the ECM .
  • Release of Growth Factors: Upon degradation of HSPGs, heparanase releases growth factors and cytokines that stimulate cell proliferation and chemotaxis .
  • Tumor Progression: Heparanase is highly expressed in various types of primary tumors, correlating with increased tumor invasiveness and vascularity .
Clinical Relevance

Due to its role in ECM degradation and tumor progression, heparanase is considered a potential target for cancer therapy. It has been implicated in promoting arterial and stent thrombosis by cleaving anticoagulant heparan sulfate proteoglycans . Additionally, heparanase interacts with resistin, influencing inflammatory conditions .

Applications in Research

Recombinant Human Heparanase-1 WB Control is used as a positive control in western blot analysis. It is prepared by using 20 μl of the recombinant protein per lane, in conjunction with monoclonal or polyclonal antibodies against heparanase . This control ensures the accuracy and reliability of western blot results.

Storage and Handling

The protein is typically stored at –20ºC to maintain its stability and activity. It is important to avoid repeated freeze-thaw cycles to prevent degradation .

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.