Heparanase Active Enzyme is produced in CHO cells.
The protein is purified by several orthogonal chromatography steps.
Heparanase Active Enzyme is identified by Western blot analysis with polyclonal
rabbit anti-HPA1 antibodies as 2 subunits of 8-kDa and 50-kDa.
Heparanase-1 (HPSE) is the only known mammalian enzyme responsible for the cleavage of heparan sulfate (HS) side chains from heparan sulfate proteoglycans (HSPGs). It plays a key role in the remodeling and degradation of the extracellular matrix (ECM) . Under normal physiological conditions, HPSE expression is tightly regulated and contributes to wound healing, embryonic development, and immune response. Its endo-β-D-glucuronidase activity specifically cleaves the β(1,4)-glycosidic bonds between glucuronic acid and glucosamine residues in HS chains .
Several methodological approaches have been developed to measure HPSE activity:
Radiolabeled substrate assays: This traditional method uses [³H]-labeled HS as substrate, with activity quantified by measuring radioactivity of [³H]-HS that remains bound to chicken histidine glycoprotein (cHRG) column after HPSE cleavage .
Immobilized substrate methods: For higher throughput, [³H]-labeled HS can be immobilized on sepharose beads or streptavidin-coated plates, allowing for detection of HPSE activity through measurement of released radioactivity .
Colorimetric-based assays: These methods rely on the specific binding of basic fibroblast growth factor (bFGF) to HS. HPSE activity is determined by measuring the reduction in bFGF binding to HS, as indicated by color development produced by TMB peroxidase .
HPSE is synthesized as a latent 65 kDa proenzyme that requires proteolytic processing to generate the active heterodimer. The active form consists of an 8 kDa subunit (Gln36–Glu109) and a 50 kDa subunit (Lys158–Ile543) that associate non-covalently .
Researchers can distinguish between these forms through:
SDS-PAGE and western blotting with antibodies specific to different domains
Size exclusion chromatography separation based on molecular weight differences
Activity assays, as only the processed heterodimeric form exhibits enzymatic activity
Analysis of glycosylation patterns, as both forms may have distinct post-translational modifications
The Protein Repair One Stop Shop (PROSS) algorithm has been successfully employed to design stable HPSE variants for bacterial expression. Unlike conventional consensus mutagenesis, PROSS combines phylogenetic sequence information with computational modeling using Rosetta forcefield-based approaches .
The implementation process involves:
Submitting the crystal structure (e.g., PDB ID: 5E9C) to the PROSS server with constraints on residues involved in substrate binding and heterodimer interface
Testing multiple design variants (typically 7 or more) with accumulated mutations
Identifying the most soluble design through expression trials
In one successful application, a HPSE variant containing 26 amino acid substitutions (HPSE P6) maintained comparable catalytic activity and identical inhibition profiles to wild-type HPSE while dramatically improving solubility and expression yield (4 mg/L in E. coli) .
Structure-activity relationship (SAR) studies have revealed critical insights for designing effective small-molecule HPSE inhibitors:
Core structure importance: Several promising scaffolds have been identified through high-throughput screening, including benzimidazole cores, isoindole-5-carboxylic acid moieties, and benzoxazole compounds .
Essential functional groups: Carboxylic acid groups are often critical for HPSE inhibition activity and cannot be modified without significant activity reduction .
Optimization pathways: Systematic modification of:
Linker groups between core structures
Substitution patterns on pendant benzyl rings
Introduction of symmetric elements (e.g., urea compounds)
Key examples include compound series with benzimidazole cores where optimization of the linker groups improved both anti-HPSE activity and pharmacokinetic profiles. For instance, a compound designated as 3f demonstrated both good HPSE inhibitory activity and favorable pharmacokinetic properties in mice, while compound 3d showed the best anti-HPSE activity with an IC₅₀ of 230 nM .
Expressing active human HPSE in prokaryotic systems presents several challenges:
Insolubility issues: Wild-type HPSE tends to form inclusion bodies when expressed in E. coli, particularly the 50 kDa subunit which typically remains completely insoluble .
Post-translational modifications: HPSE contains glycosylation sites and disulfide bonds that are difficult to replicate in bacterial systems .
Proper heterodimer formation: The active enzyme requires correct association of the 8 kDa and 50 kDa subunits.
These challenges can be overcome through:
Computational redesign: Using PROSS algorithm to introduce stability-enhancing mutations while preserving function .
Specialized expression strains: Employing E. coli Shuffle T7 Express cells that allow disulfide bond formation in the cytoplasm .
Co-expression with chaperones: Trigger factor and GroEL/GroES chaperones help with proper protein folding .
Dual expression vectors: Using vectors like pETDuet-1 for coordinated expression of both subunits .
Optimized purification: Sequential purification using Ni²⁺-NTA, heparin, and size exclusion chromatography to obtain pure, homogeneous, heterodimeric HPSE .
Small-molecule HPSE inhibitors and heparan sulfate (HS) mimetics differ significantly in their inhibitory mechanisms and pharmacological properties:
Binding mechanism:
Pharmacokinetic properties:
Target selectivity:
Many HS mimetics lack specificity and can interact with other heparin-binding proteins
Well-designed small-molecule inhibitors can achieve higher selectivity, as exemplified by compound 6b (4-Bn-RK-682), which demonstrated selective HPSE inhibition without off-target effects on protein tyrosine phosphatases
Several high-throughput screening (HTS) strategies have proven effective for identifying novel HPSE inhibitors:
Radiometric assays: Using [³H]-labeled HS immobilized on sepharose beads or streptavidin-coated plates to detect inhibition of HPSE activity through measurement of retained radioactivity .
Colorimetric assays: Based on specific binding of basic FGF (bFGF) to HS, measuring reduction in bFGF binding as indicated by color development from TMB peroxidase .
Virtual screening approaches: Computational methods exploiting the available crystal structures of HPSE to identify potential inhibitors through molecular docking and structure-based design .
The effectiveness of these approaches is demonstrated by the discovery of several lead compounds:
Benzimidazole core lead compound 1 was identified through radiometric HTS
Isoindole-5-carboxylic acid/benzoxazole compound 7 (IC₅₀: 8 μM) was discovered using colorimetric-based HTS
Furanylthiazole acetic acid lead compound 12 (IC₅₀: 25 μM) was identified and optimized to yield compounds with IC₅₀ values as low as 300-400 nM
The computational design of stable HPSE variants has revealed important structure-function correlations:
Mutation distribution patterns: Stability-enhancing mutations are predominantly located on the protein surface, away from the active site and heterodimer interface .
Structural conservation: Despite introducing 26 mutations in the HPSE P6 variant, the enzyme remained essentially structurally isomorphous to wild-type HPSE, with:
Dynamic properties: Molecular dynamics simulations confirmed that the engineered variants exhibited dynamics largely identical to wild-type enzyme, indicating no significant changes to the relative stabilities of different conformational substates .
Functional neutrality: The mutations acquired through phylogenetic analysis reinforced the concept of functional neutrality of many surface residues, as the engineered HPSE variant maintained:
Multidimensional data tables offer powerful tools for analyzing complex relationships between multiple variables affecting HPSE activity:
Application scenarios: This approach is particularly valuable for HPSE research when investigating:
Implementation approach:
Advantage over multiple simple tables: A single multidimensional data table allows researchers to examine interactions between three or more variables simultaneously, providing more comprehensive insights than could be achieved with multiple two-dimensional tables .
Detecting low levels of HPSE activity in biological samples requires highly sensitive methodologies:
Fluorogenic substrate assays: Using synthetic substrates with fluorescent tags that are released upon HPSE cleavage, providing real-time monitoring capabilities with nanomolar sensitivity.
ELISA-based detection systems: Combining substrate-specific antibodies with enzyme-linked detection for quantification of HPSE-cleaved products in complex biological matrices.
Mass spectrometry approaches: Analyzing the specific fragmentation patterns of HS oligosaccharides produced by HPSE digestion, allowing for both quantification and structural characterization.
Enhanced radiometric methods: Modifications of traditional [³H]-labeled HS assays with improved signal amplification and background reduction for detecting extremely low enzyme concentrations.
A comprehensive validation approach for novel HPSE inhibitors should include:
In vitro enzyme inhibition characterization:
Cell-based assays:
Comparative analysis with reference compounds:
Structural characterization of inhibitor binding:
Animal model validation:
Several emerging technologies hold promise for advancing HPSE inhibitor development:
AI-driven drug design: Machine learning algorithms trained on existing HPSE inhibitor data could identify novel chemical scaffolds and predict activity with greater efficiency than traditional screening approaches.
Fragment-based drug discovery (FBDD): This approach can identify low molecular weight fragments that bind to different regions of HPSE, which can then be linked or grown to create highly specific inhibitors with improved drug-like properties.
Cryo-electron microscopy: Advancements in cryo-EM resolution may enable visualization of HPSE in different conformational states, providing insights into dynamics that X-ray crystallography cannot capture.
Activity-based protein profiling: Development of activity-based probes specific for HPSE could enable direct assessment of inhibitor engagement with active enzyme in complex biological systems.
Targeted protein degradation: Adapting PROTAC (Proteolysis Targeting Chimera) technology to HPSE could provide an alternative approach to functional inhibition by inducing selective degradation of the enzyme.
Experimental approaches to studying HPSE activity require customization based on disease context:
Cancer research:
Inflammatory diseases:
Emphasis on immune cell recruitment and activation
Analysis of cytokine release and signaling
Evaluation of tissue-specific inflammation models
Assessment of HS degradation products as inflammation mediators
Diabetes and metabolic disorders:
Focus on vascular complications and endothelial dysfunction
Analysis of basement membrane integrity
Evaluation of kidney filtration barrier function
Assessment of interaction with glucose metabolism pathways
Viral infections:
Investigation of viral attachment and entry mechanisms
Analysis of immune evasion strategies
Evaluation of viral replication in presence of HPSE inhibition
Assessment of potential as broad-spectrum antiviral approach
Compound Class | Representative Example | IC₅₀ Value | Key Structural Features | Additional Activities | Development Stage |
---|---|---|---|---|---|
Benzimidazole derivatives | Compound 3d | 230 nM | Benzimidazole core with optimized linker groups | Not reported | Preclinical |
Symmetric urea compounds | Compounds 4a-4g | Various | Symmetric structure with dual binding | Not reported | Preclinical |
RK-682 derivatives | Compound 6b (4-Bn-RK-682) | Similar to RK-682 | Selective for HPSE | Inhibition of invasion and metastasis in HT1080 cells | Preclinical |
Isoindole carboxylic acid/benzoxazole | Compounds 9n, 11c | 200 nM | Carboxylic acid essential for activity | Not reported | Preclinical |
Furanylthiazole acetic acid | Compounds 14b, 14e | 400 nM | Furanylthiazole core | Not reported | Preclinical |
Benzoxazole acetic acid | Compounds 17j, 18g | 300 nM | Benzoxazole acetic acid core | Anti-angiogenesis activity | Preclinical |
Research tool compounds | Compound 16d (OGT2115) | Not specified | Not fully described | Used as research tool | Research use only |
Recombinant Human Heparanase-1 is expressed in human HEK 293 cells as an active heterodimer glycoprotein containing a 50 kDa subunit and an 8 kDa subunit . This expression system allows human-like glycosylation and folding, which often supports higher specific activity of the protein . The protein is produced without artificial tags, ensuring its functionality and stability.
Heparanase-1 is the only heparan sulfate-degrading endoglycosidase. It cleaves the heparan sulfate side chains of heparan sulfate proteoglycans into fragments of 10–20 sugar units . This enzyme is initially translated as a preproenzyme containing a signal peptide. The cleavage of the signal sequence yields a latent 65 kDa pro-heparanase, which must undergo further processing to become active. The mature active heparanase is a heterodimer consisting of an N-terminal 8 kDa subunit and a C-terminal 50 kDa subunit .
Heparanase-1 plays a crucial role in the regulation of multiple biological processes, including tumor growth, metastasis, angiogenesis, and inflammation . It is involved in extracellular matrix (ECM) remodeling and the release of heparan sulfate-linked biological molecules, such as cytokines and growth factors . The enzyme’s activity is measured by its ability to cleave heparan sulfate from substrates like Syndecan-4 .
Due to its involvement in critical biological processes, recombinant human heparanase-1 is extensively used in research to study cancer progression, tissue regeneration, and inflammatory diseases . Its ability to degrade heparan sulfate makes it a valuable tool for investigating the ECM’s role in various physiological and pathological conditions.
The recombinant human heparanase-1 is produced with high purity, typically greater than 95%, as determined by SDS-PAGE . It is supplied as a lyophilized powder or in a solution, depending on the manufacturer. The endotoxin level is kept below 1.0 EU per 1 μg of protein to ensure its suitability for sensitive biological applications .