HS3ST1 Human

Heparan Sulfate 3-O-Sulfotransferase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Alzheimer’s Disease (AD):

  • HS3ST1 is a genetic risk locus identified in genome-wide association studies (GWAS) involving 9,751 patients .

  • Overexpression in AD patients correlates with a sevenfold increase in 3-O-sulfated HS in brain tissues, accelerating tau protein internalization and pathology .

  • Knockout mouse studies confirm that 3-O-sulfated HS produced by HS3ST1 facilitates tau spreading .

Atherosclerosis and Inflammation:

  • An intronic SNP (rs16881446) in HS3ST1 reduces endothelial gene expression, increasing susceptibility to coronary artery disease .

  • Hs3st1<sup>−/−</sup> mice exhibit heightened sensitivity to LPS-induced septic shock due to dysregulated anti-inflammatory activity of antithrombin .

Other Associations:

  • Polymorphisms linked to altered heparan sulfate structures in colorectal cancer and schizophrenia .

Tau Pathology in AD (2023):

  • Method: LC-MS/MS analysis of 44 human brain samples .

  • Finding: 3-O-sulfated HS (Tetra-1 domain) increased sevenfold in AD brains (P < 0.0005) .

  • Mechanism: Synthetic 3-O-sulfated tetradecasaccharides enhance tau internalization, suggesting HS3ST1-driven HS modifications promote neurofibrillary tangle formation .

Sepsis and Inflammation (2017):

  • Model: Hs3st1<sup>−/−</sup> mice challenged with LPS .

  • Result: Increased mortality (hazard ratio: 4.42) due to TNF-α hypersensitivity .

  • Implication: HS<sup>AT+</sup> regulates antithrombin’s anti-inflammatory activity .

StudyKey FindingDisease Relevance
LC-MS/MS brain analysis3-O-sulfated HS elevation drives tau uptakeAlzheimer’s disease
Hs3st1<sup>−/−</sup> miceLoss of HS<sup>AT+</sup> increases inflammatory mortalitySepsis, atherosclerosis
GWAS meta-analysisHS3ST1 variants associate with AD and dyslipidemiaNeurodegeneration

Therapeutic Implications

  • Targeting HS3ST1: Inhibition of 3-O-sulfotransferase activity may slow tau propagation in AD .

  • Biomarker Potential: Elevated 3-O-sulfated HS levels in cerebrospinal fluid could aid early AD diagnosis .

Research Gaps and Future Directions

  • Structural Biology: Detailed enzyme-substrate interaction studies are needed to design isoform-specific inhibitors .

  • Longitudinal Studies: Validate HS3ST1’s role in tauopathy progression across diverse populations .

Product Specs

Introduction
Heparan Sulfate 3-O-Sulfotransferase 1 (HS3ST1) is an enzyme that facilitates the transfer of a sulfo group from a molecule called 3'-phospho-5'-adenylyl sulfate (PAPS) to a specific position (position 3) on glucosamine units within heparan. This enzymatic activity is critical in the production of a molecule called heparan sulfate (HSact), particularly in making it functional in preventing blood clotting. The significance of HS3ST1 lies in its role in forming the binding site for antithrombin, a key molecule involved in blood clotting regulation.
Description
This product consists of the human form of HS3ST1, a protein produced in E. coli bacteria. It is a single chain of 310 amino acids (specifically amino acids 21 to 307), without any sugar modifications, and has a molecular weight of 36.2 kilodaltons. For purification and detection purposes, a 23 amino acid tag called His-tag is attached to the protein's beginning. The purification process involves specialized chromatography techniques.
Physical Appearance
The product is a clear, colorless solution that has been sterilized through filtration.
Formulation

The product is provided as a solution of HS3ST1 protein at a concentration of 0.25 milligrams per milliliter. The solution is buffered with 20mM Tris-HCl at a pH of 8.0 and also contains 0.2M NaCl, 40% glycerol, and 2mM DTT.

Stability
For short-term storage (up to 2-4 weeks), the product can be kept at 4 degrees Celsius. For extended storage, freezing at -20 degrees Celsius is recommended. To further enhance stability during long-term storage, consider adding a carrier protein like HSA or BSA at a concentration of 0.1%. It's important to avoid repeatedly freezing and thawing the product.
Purity
The purity of the HS3ST1 protein in this product is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms
Heparan sulfate glucosamine 3-O-sulfotransferase 1, Heparan sulfate D-glucosaminyl 3-O-sulfotransferase 1, 3-OST-1, Heparan sulfate 3-O-sulfotransferase 1,
h3-OST-1, HS3ST1, 3OST, 3OST1, HS3S1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSRPAELGQ QELLRKAGTL QDDVRDGVAP NGSAQQLPQT IIIGVRKGGT RALLEMLSLH PDVAAAENEV HFFDWEEHYS HGLGWYLSQM PFSWPHQLTV EKTPAYFTSP KVPERVYSMN PSIRLLLILR DPSERVLSDY TQVFYNHMQK HKPYPSIEEF LVRDGRLNVD YKALNRSLYH VHMQNWLRFF PLRHIHIVDG DRLIRDPFPE IQKVERFLKL SPQINASNFY FNKTKGFYCL RDSGRDRCLH ESKGRAHPQV DPKLLNKLHE YFHEPNKKFF ELVGRTFDWH.

Q&A

What is HS3ST1 and what is its primary function in human cells?

HS3ST1 is an enzyme that catalyzes the 3-O-sulfation of heparan sulfate (HS) proteoglycans. Located on chromosome 4 in humans, this gene encodes the 3-O-sulfotransferase isoform 1 (3-OST-1) . The primary function of HS3ST1 is to modify heparan sulfate by adding a sulfate group at the 3-O position of glucosamine residues, creating specific sulfation patterns crucial for various biological interactions. One of the most well-characterized functions of 3-O-sulfated HS is its interaction with antithrombin (AT), which plays a key role in anticoagulation and anti-inflammatory processes .

Methodologically, researchers can study HS3ST1 function through various approaches, including recombinant enzyme assays with defined HS substrates, genetic knockout models, and analysis of 3-O-sulfated HS structures using liquid chromatography-mass spectrometry (LC-MS/MS) .

How is the expression of HS3ST1 dysregulated in pathological conditions?

HS3ST1 expression shows significant dysregulation across multiple pathological conditions:

  • In non-small-cell lung cancer (NSCLC), HS3ST1 expression is significantly upregulated in tumor tissues compared to adjacent normal tissues . This overexpression promotes malignant behaviors including enhanced proliferation, reduced apoptosis, and increased migration capabilities.

  • In Alzheimer's disease (AD), HS3ST1 is overexpressed as a genetic risk gene, resulting in a sevenfold increase of a specific 3-O-sulfated HS structure in AD brain samples (p < 0.0005) .

  • In atherosclerosis, certain HS3ST1 genotypes (particularly the rs16881446 G/G genotype) associate with reduced HS3ST1 expression in endothelial cells, which correlates with increased severity of coronary artery disease and higher rates of atherosclerotic cardiovascular events .

These expression patterns suggest that HS3ST1 may serve as both a biomarker and therapeutic target across multiple disease contexts.

What techniques are most effective for analyzing HS3ST1 expression and function in experimental models?

Several complementary techniques provide robust analysis of HS3ST1 expression and function:

TechniqueApplicationAdvantagesLimitations
Quantitative PCR (qPCR)mRNA expression measurementHigh sensitivity, quantitativeDoes not assess protein levels or activity
Western blottingProtein expression analysisDirectly measures protein levelsSemi-quantitative, antibody dependent
LC-MS/MSAnalysis of 3-O-sulfated HS structuresPrecise structural characterizationRequires specialized equipment, complex analysis
siRNA/vector transfectionModulation of HS3ST1 expressionAllows functional studiesTransient effects, potential off-target impacts
CRISPR-Cas9Gene knockout/editingPermanent modification, highly specificComplex delivery, potential compensatory mechanisms
In vivo mouse modelsPhysiological relevanceWhole-organism effectsSpecies differences, complex phenotypes

For cell-based models, researchers have successfully employed lentiviral vectors for HS3ST1 overexpression and siRNA for knockdown in lung cancer cell lines . The efficacy of these manipulations should be confirmed through qPCR and Western blot analysis before proceeding to functional assays.

How can researchers effectively analyze the specific 3-O-sulfated HS structures generated by HS3ST1?

Analysis of specific 3-O-sulfated HS structures requires specialized approaches due to their structural complexity:

  • LC-MS/MS represents the gold standard for detailed structural analysis of HS sulfation patterns. This approach has successfully identified a specific 3-O-sulfated HS structure significantly elevated in Alzheimer's disease brain samples .

  • Comparative analysis using recombinant sulfotransferases to generate defined structures provides valuable reference standards. This approach, combined with HS from genetic knockout mice as controls, helps validate structural assignments .

  • For functional studies, synthetic oligosaccharides carrying specific 3-O-sulfated domains (such as the tetradecasaccharide/14-mer used in AD research) allow precise structure-function relationships to be established .

  • Binding assays with proteins known to interact with 3-O-sulfated HS (such as antithrombin) provide indirect but functionally relevant assessment of specific sulfation patterns.

When designing experiments, researchers should consider incorporating multiple complementary approaches to overcome the limitations of individual methods.

What mechanisms underlie HS3ST1's role in cancer progression?

HS3ST1 promotes cancer progression through multiple interconnected mechanisms, particularly well-documented in non-small-cell lung cancer (NSCLC):

  • Cell proliferation and survival: HS3ST1 overexpression significantly enhances NSCLC cell proliferation while reducing apoptosis. At the molecular level, HS3ST1 decreases expression of pro-apoptotic proteins (cleaved-caspase-8, cleaved-caspase-3, and Bax) while increasing anti-apoptotic protein Bcl-xl expression .

  • Cell cycle regulation: HS3ST1 overexpression reduces the proportion of NSCLC cells in G0/G1 phase, promoting cell cycle progression .

  • NF-κB pathway activation: Mechanistically, HS3ST1 inhibits spot-type zinc finger protein (SPOP) expression, which normally mediates the degradation of Fas-associated death domain protein (FADD). This inhibition leads to activation of the NF-κB pathway, promoting tumor cell survival and proliferation .

  • Tumor growth in vivo: Studies in mouse models have confirmed that HS3ST1 overexpression enhances tumor growth, while HS3ST1 knockdown significantly attenuates tumor development .

These findings suggest that targeting HS3ST1 could potentially inhibit cancer progression through multiple mechanistic pathways.

How does HS3ST1 contribute to Alzheimer's disease pathogenesis?

HS3ST1 contributes to Alzheimer's disease (AD) pathogenesis through its role in tau pathology propagation:

  • Genetic risk: HS3ST1 has been identified as a genetic risk gene associated with AD, with evidence of overexpression in patient samples .

  • Specific 3-O-sulfated HS: LC-MS/MS analysis of brain samples has revealed that a specific 3-O-sulfated HS structure displays a sevenfold increase in AD brains compared to controls (n = 14, P < 0.0005). This specific structure is produced by 3-OST-1, encoded by the HS3ST1 gene .

  • Enhanced tau pathology: Mechanistically, the specific 3-O-sulfated HS domain facilitates tau cellular uptake. Experimental evidence shows that a synthetic tetradecasaccharide (14-mer) carrying the specific 3-O-sulfated domain exhibits stronger inhibition of tau internalization compared to a 14-mer without this domain, suggesting it competes with cellular 3-O-sulfated HS that mediates tau uptake .

  • Potential therapeutic target: These findings indicate that HS3ST1 overexpression may enhance the spread of tau pathology in AD, positioning it as a novel therapeutic target by potentially reducing tau propagation throughout the brain .

Researchers studying HS3ST1 in AD should consider both genetic approaches (examining polymorphisms and expression patterns) and structural analyses of the specific 3-O-sulfated HS domains involved in tau interactions.

What is the connection between HS3ST1 genotypes and inflammatory vascular diseases?

HS3ST1 genotypes significantly impact inflammatory vascular diseases, particularly atherosclerosis, through modulation of antithrombin's (AT) anti-inflammatory activities:

  • Genetic association: A candidate-gene association study involving over 2,000 coronary catheterization patients identified an intronic SNP, rs16881446, in a putative transcriptional regulatory region of the HS3ST1 gene. The rs16881446 G/G genotype independently associated with increased severity of coronary artery disease and higher rates of atherosclerotic cardiovascular events .

  • Reduced HS3ST1 expression: In primary endothelial cells, the rs16881446 G allele associated with decreased HS3ST1 expression. This reduction results in decreased production of the specific 3-O-sulfated heparan sulfate that mediates AT's anti-inflammatory activity (HS AT+) .

  • Altered vascular responses: Experimental studies in Hs3st1 knockout mice demonstrated that without sufficient HS AT+, AT's pro-inflammatory effects predominate. When treated with AT, Hs3st1+/+ mice exhibited reduced leukocyte adhesion to endothelial cells and dilated coronary arterioles, indicating anti-inflammatory effects. In contrast, Hs3st1-/- mice showed opposite responses to AT treatment .

  • Sepsis susceptibility: Beyond atherosclerosis, Hs3st1-/- mice showed increased susceptibility to LPS-induced death due to heightened sensitivity to TNF, further demonstrating HS3ST1's role in regulating inflammatory responses .

These findings establish HS3ST1 as a critical regulator of vascular inflammation through its generation of specific 3-O-sulfated HS domains that mediate AT's anti-inflammatory activity.

How do the signaling pathways downstream of HS3ST1 interact with disease-specific molecular mechanisms?

HS3ST1 influences multiple signaling pathways with distinct implications across different disease contexts:

  • NF-κB pathway in cancer: In NSCLC, HS3ST1 promotes malignancy primarily through inhibition of SPOP, which normally mediates FADD degradation. This inhibition leads to NF-κB pathway activation, promoting cancer cell survival and proliferation . Researchers should investigate whether this mechanism extends to other cancer types and whether it interacts with tumor-specific oncogenic pathways.

  • Tau pathology in Alzheimer's disease: The specific 3-O-sulfated HS structure generated by HS3ST1 enhances tau cellular uptake, potentially accelerating the spread of tau pathology in AD . Future research should explore the molecular interactions between 3-O-sulfated HS and tau, including potential binding sites and conformational changes that facilitate internalization.

  • Anti-inflammatory signaling in vascular disease: In endothelial cells, HS3ST1-generated 3-O-sulfated HS mediates antithrombin's anti-inflammatory activity, reducing leukocyte adhesion and promoting vasodilation . The precise signaling events downstream of AT binding to 3-O-sulfated HS remain to be fully elucidated but likely involve altered expression of adhesion molecules and inflammatory cytokines.

Methodologically, researchers can employ phosphoproteomic analysis, transcriptomics, and protein-protein interaction studies after HS3ST1 modulation to map these signaling networks comprehensively. Integration of multi-omics data will be essential to understand context-specific signaling outcomes.

What approaches can resolve contradictory findings regarding HS3ST1 function across different experimental systems?

Several methodological approaches can help resolve apparent contradictions in HS3ST1 research:

  • Context-specific analysis: HS3ST1 exhibits different, sometimes opposing effects depending on the cellular context. For example, it shows pro-tumorigenic effects in NSCLC but potentially protective effects in vascular inflammation through its impact on AT signaling . Researchers should carefully document cell type, disease context, and experimental conditions when reporting findings.

  • Structural specificity: The specific 3-O-sulfated HS structures generated by HS3ST1 may vary across cell types and conditions. Using LC-MS/MS to characterize these structures in each experimental system can help explain functional differences .

  • Genetic background consideration: Studies in mouse models have shown that Hs3st1 knockout produces different responses to inflammatory stimuli compared to wild-type mice . Researchers should consider genetic background, including potential compensatory mechanisms, when interpreting knockout studies.

  • Temporal dynamics: The effects of HS3ST1 may vary depending on acute versus chronic conditions. For example, its role in acute inflammation versus chronic diseases like atherosclerosis may involve different mechanisms . Time-course experiments can help clarify these dynamics.

  • Pathway interactions: HS3ST1's effects are mediated through interactions with multiple pathways, including NF-κB signaling and AT activity. Comprehensive pathway analysis, rather than focusing on isolated endpoints, can help reconcile apparently contradictory findings.

How might novel synthetic biology approaches advance HS3ST1 research?

Synthetic biology offers promising approaches to overcome current limitations in HS3ST1 research:

  • Engineered HS sulfation patterns: CRISPR-based editing of multiple sulfotransferase genes could generate cell lines with defined HS sulfation patterns, allowing precise structure-function studies. This approach would overcome the heterogeneity of naturally occurring HS structures that complicates interpretation of experimental results.

  • Optogenetic regulation of HS3ST1: Developing light-inducible HS3ST1 expression systems would enable temporal control of enzyme activity, facilitating studies of acute versus chronic effects and helping to resolve contradictory findings that may depend on timing of expression.

  • Synthetic oligosaccharide libraries: Expanding on the tetradecasaccharide approach used in AD research , creating libraries of synthetic HS oligosaccharides with defined sulfation patterns would enable systematic screening for structure-function relationships across different disease contexts.

  • Cell-specific HS3ST1 modulation in vivo: Using tissue-specific promoters to drive Cre recombinase expression for conditional Hs3st1 knockout in mouse models would help distinguish cell-autonomous effects from systemic impacts of HS3ST1 activity, particularly important in complex diseases involving multiple cell types.

  • Biosensors for 3-O-sulfated HS: Developing fluorescent or bioluminescent reporters that specifically recognize 3-O-sulfated HS domains would enable real-time monitoring of HS3ST1 activity in living cells and tissues, providing insights into the dynamics of HS modification in response to different stimuli.

What therapeutic strategies targeting HS3ST1 show the most promise for clinical development?

Several therapeutic strategies targeting HS3ST1 show potential for clinical development:

Therapeutic ApproachDisease ApplicationMechanismDevelopment Status
Small molecule HS3ST1 inhibitorsCancer (NSCLC)Reduce 3-O-sulfation to inhibit pro-tumorigenic pathwaysPreclinical research
RNAi-based HS3ST1 suppressionCancerReduce HS3ST1 expression to inhibit tumor growthPreclinical research
Synthetic 3-O-sulfated HS mimeticsAlzheimer's diseaseCompetitively inhibit tau-HS interactions to reduce pathology spreadEarly research
Recombinant AT derivativesAtherosclerosisEnhance anti-inflammatory effects in patients with HS3ST1 risk genotypesConceptual
Antibodies targeting specific 3-O-sulfated HS domainsCancer, neurodegenerationBlock disease-promoting interactions of specific HS domainsEarly research

For cancer applications, targeting HS3ST1's effects on the SPOP/FADD/NF-κB pathway represents a promising approach, as this mechanism has been well-validated in NSCLC models . For Alzheimer's disease, synthetic oligosaccharides that competitively inhibit interactions between tau and 3-O-sulfated HS could potentially reduce tau propagation, as suggested by experimental data with the tetradecasaccharide (14-mer) .

The development of these therapeutic approaches requires addressing several challenges, including achieving specificity for HS3ST1 versus other sulfotransferases, developing effective delivery methods for targeting specific tissues, and understanding potential compensatory mechanisms that might limit efficacy.

What biomarker strategies based on HS3ST1 could improve disease diagnosis and monitoring?

HS3ST1-based biomarker strategies offer potential advances for multiple diseases:

  • Genetic biomarkers: Genotyping for HS3ST1 variants such as rs16881446 could identify individuals at higher risk for atherosclerotic cardiovascular events , enabling preventive interventions in high-risk populations. This approach requires minimal invasiveness (blood sampling) and established genotyping technologies.

  • Specific 3-O-sulfated HS structures: The sevenfold increase in specific 3-O-sulfated HS structures in AD brain samples suggests potential as a disease biomarker. Research should focus on detecting these structures in more accessible biofluids (CSF, plasma) using targeted LC-MS/MS approaches.

  • HS3ST1 expression levels: Measuring HS3ST1 expression in tumor biopsies could potentially serve as a prognostic or predictive biomarker in cancers like NSCLC, where overexpression correlates with more aggressive disease . Standardized IHC or qPCR protocols would need development and validation.

  • Pathway activation markers: Downstream markers of HS3ST1 activity, such as NF-κB pathway activation or changes in apoptotic markers (Bax, Bcl-xl, cleaved caspases), could serve as surrogate biomarkers in cancer applications .

  • Integrative multi-biomarker panels: Combining HS3ST1 genetic variants, expression levels, and downstream pathway markers could provide more robust disease prediction than any single marker alone.

Implementation challenges include standardizing detection methods for complex HS structures, validating biomarkers across diverse patient populations, and establishing clinically meaningful cutoff values for diagnostic or prognostic applications.

How might computational approaches enhance prediction of HS3ST1-mediated disease mechanisms?

Advanced computational approaches offer significant potential for advancing HS3ST1 research:

  • Molecular dynamics simulations: Computational modeling of 3-O-sulfated HS interactions with proteins like antithrombin and tau can predict binding sites, conformational changes, and interaction energies, helping prioritize experimental studies. These simulations can incorporate atomic-level detail of specific sulfation patterns generated by HS3ST1.

  • Machine learning for structure-function prediction: Training algorithms on existing data linking HS structures to biological functions could enable prediction of novel functions for specific 3-O-sulfated domains. This approach could help identify previously unrecognized roles for HS3ST1 in different disease contexts.

  • Network analysis of multi-omics data: Integration of genomics, transcriptomics, proteomics, and glycomics data through network analysis can reveal context-specific HS3ST1 functions and identify key nodes in HS3ST1-regulated pathways. This could uncover potential therapeutic targets beyond HS3ST1 itself.

  • Genome-wide association study (GWAS) data mining: Systematic analysis of existing GWAS datasets for associations with HS3ST1 variants beyond the already identified rs16881446 SNP could reveal additional disease connections not yet experimentally validated.

  • Pharmacophore modeling: Computational approaches to design small molecules targeting HS3ST1 or specific 3-O-sulfated HS structures could accelerate therapeutic development. Virtual screening of compound libraries against these models could identify lead candidates for experimental testing.

These computational approaches are most powerful when integrated with experimental validation, creating an iterative cycle of prediction, testing, and refinement to advance understanding of HS3ST1's complex roles in human disease.

Product Science Overview

Introduction

Heparan Sulfate 3-O-Sulfotransferase 1 (HS3ST1) is an enzyme that plays a crucial role in the biosynthesis of heparan sulfate, a linear polysaccharide found in the extracellular matrix and on cell surfaces. This enzyme is responsible for the transfer of a sulfo group to the 3-position of glucosamine residues in heparan sulfate, a modification that is essential for the biological activity of heparan sulfate .

Structure and Function

HS3ST1 is a member of the heparan sulfate biosynthetic enzyme family. It possesses both heparan sulfate glucosaminyl 3-O-sulfotransferase activity and anticoagulant heparan sulfate conversion activity . The enzyme uses 3’-phospho-5’-adenylyl sulfate (PAPS) as a sulfo group donor to catalyze the transfer of the sulfo group . This reaction is considered the rate-limiting step in the biosynthesis of anticoagulant heparan sulfate .

Biological Significance

Heparan sulfate proteoglycans (HSPGs) are involved in various physiological processes, including embryogenesis, angiogenesis, blood coagulation, and inflammation . The 3-O-sulfation of heparan sulfate by HS3ST1 is a rare but critical modification that influences the binding of heparan sulfate to a variety of proteins, including growth factors, cytokines, and extracellular matrix components . These interactions can affect cellular processes such as proliferation, migration, apoptosis, and immune response .

Clinical Relevance

Alterations in the expression of HS3ST1 have been observed in various cancers . Dysregulation of this enzyme can lead to significant changes in the structure of heparan sulfate, impacting tumorigenesis and cancer progression . The enzyme’s role in modifying heparan sulfate also makes it a potential target for therapeutic strategies in cancer treatment .

Recombinant HS3ST1

Recombinant human HS3ST1 is produced using in vitro expression systems, such as wheat germ expression systems . The recombinant protein typically includes a GST tag at the N-terminal and corresponds to the amino acids 1-307 of the full-length human HS3ST1 . This recombinant form is used in various research applications, including Western blotting, ELISA, and protein array .

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.