CHST11 (Carbohydrate Sulfotransferase 11), also known as chondroitin 4-O-sulfotransferase 1 (C4ST-1), is an enzyme that catalyzes the transfer of sulfate to position 4 of the N-acetylgalactosamine (GalNAc) residue of chondroitin. It belongs to the sulfotransferase 2 family and is localized to the Golgi membrane .
Functionally, CHST11:
Contributes to the synthesis of chondroitin sulfate, a predominant proteoglycan in cartilage
Is widely expressed across tissues, with particularly high expression in spleen, thymus, bone marrow, peripheral blood leukocytes, lymph node, heart, brain, lung, and placenta
Can also sulfate galactose residues in desulfated dermatan sulfate
Shows preference for sulfating GlcA->GalNAc units over IdoA->GalNAc units
Recent research indicates that CHST11 plays significant roles in cancer progression, particularly in hepatocellular carcinoma and breast cancer, through modulation of tumor microenvironments and immune cell infiltration .
CHST11 antibodies have several important applications in research settings:
Western Blot (WB): For detecting and quantifying CHST11 protein expression in cellular lysates. Most commercial antibodies are validated for WB with dilution recommendations between 1:500-2000 .
Immunohistochemistry (IHC): For examining CHST11 expression patterns in tissue samples. In particular, CHST11 protein expression has been assessed by IHC staining in HCC and adjacent normal liver specimens at 1:100 dilution .
Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative detection of CHST11, with typical recommended dilutions of 1:5000-20000 .
Immunofluorescence (IF): For visualizing subcellular localization of CHST11, particularly when studying its relationship with other proteins like E-cadherin and β-catenin in cancer cells undergoing epithelial-mesenchymal transition .
Flow Cytometry: For analyzing CHST11 expression in individual cells, particularly when studying its relationship with cell surface markers .
Ensuring antibody specificity is critical for meaningful research. For CHST11 antibodies, the following validation approaches are recommended:
Positive and Negative Controls:
Use tissue/cell types known to express high levels of CHST11 (spleen, thymus, bone marrow) as positive controls
Use CHST11-knockout or siRNA-treated cells as negative controls
In research by Xiong et al., CHST11 was knocked down in Huh7 and Hep3B cells using lentivirus (si-81566 and si-81567), providing excellent negative controls for antibody validation
Western Blot Analysis:
Immunoreactive Score (IRS) Validation:
RNA-Protein Correlation:
CHST11 plays a significant role in shaping the tumor microenvironment (TME) and modulating immune cell infiltration, particularly in hepatocellular carcinoma:
Immune and Stromal Cell Infiltration:
Regulatory T Cell (Treg) Infiltration:
CHST11 expression facilitates Treg infiltration in HCC
The infiltration of Tregs is significantly higher in high-CHST11 expression groups (p = 0.018)
This correlation has been validated using multiple algorithms: CIBERSORT (R = 0.163, p = 0.002), CIBERSORT-ABS (R = 0.383, p < 0.0001), and Quantiseq (R = 0.466, p < 0.0001)
Treg Cell Markers:
Immune Checkpoint Regulation:
Methodological Approach to Study This Relationship:
Researchers should first categorize samples into high and low CHST11 expression groups
Use CIBERSORT algorithm to assess immune cell infiltration
Validate findings with multiple algorithms (TIMER2, Quantiseq)
Correlate CHST11 expression with specific immune cell markers using RNA-seq or IHC
These findings suggest that CHST11 contributes to immunosuppressive tumor microenvironments, offering potential for combination immunotherapy targeting both CHST11 and immune checkpoints.
CHST11 expression has significant prognostic implications in several cancer types:
Hepatocellular Carcinoma (HCC):
Univariate and multivariate COX analyses have identified CHST11 mRNA as an independent prognostic biomarker in HCC
Higher CHST11 expression correlates with worse survival outcomes
In Treg-enriched HCC patients, high CHST11 expression indicated particularly adverse prognosis (HR = 1.91, 95% CI = 1.22–3, p = 0.0039)
HCC patients with TP53 mutations show significantly higher CHST11 expression (p < 0.0001)
Breast Cancer:
Elevated expression of CHST11 in breast tumor specimens is significantly associated with poor survival
CHST11 expression is higher in aggressive breast cancer cell lines compared to less aggressive ones
CHST11 expression correlates with the development of epithelial-mesenchymal transition (EMT) characteristics and stem cell-like properties
Other Cancers:
Clinical Parameters Correlation:
To properly assess the prognostic value of CHST11:
Apply both univariate and multivariate COX analyses
Use Kaplan-Meier survival analysis stratified by CHST11 expression levels
Consider the interaction with immune cell infiltration, particularly Tregs
Validate findings across multiple patient cohorts
CHST11 influences cancer progression through several interconnected molecular mechanisms:
Cell Proliferation and Metastasis:
Epithelial-Mesenchymal Transition (EMT):
In breast cancer, MCF-7 cells with stable CHST11 expression (MCF-7-CHST11) display morphological characteristics consistent with EMT
These cells exhibit decreased E-cadherin expression and increased β-catenin accumulation
CHST11 overexpression results in upregulation of key EMT and stem cell markers
Wnt Signaling Pathway:
Immunosuppression Mechanisms:
Relation to TP53 Mutation:
Experimental approaches to investigate these mechanisms:
siRNA-mediated knockdown of CHST11 followed by functional assays
Gene expression analysis after CHST11 modulation
Pathway inhibition studies (e.g., Wnt inhibitors)
Co-immunoprecipitation to identify protein interaction partners
ChIP assays to determine transcriptional regulation
Experimental Approaches for CHST11 Silencing:
siRNA Transfection:
Lentiviral shRNA Delivery:
CRISPR-Cas9 Gene Editing:
For complete knockout studies, CRISPR-Cas9 targeting of CHST11 provides definitive loss-of-function
Guide RNA design should target early exons to ensure complete protein disruption
Validation of Knockdown Efficiency:
RT-qPCR to confirm reduction in CHST11 mRNA levels
Western blot using validated CHST11 antibodies to verify protein reduction
Functional assays to confirm loss of chondroitin 4-sulfotransferase activity
Observed Functional Consequences:
Cell Proliferation Effects:
Metastatic Potential:
Immune-Related Changes:
Reduced expression of immune checkpoint molecules
Altered tumor microenvironment with potential decrease in Treg infiltration
These effects can be assessed using co-culture systems with immune cells or in vivo models
Reversal of EMT Phenotype:
Wnt Signaling Modulation:
Decreased β-catenin accumulation and reduced Wnt target gene expression
These effects can be measured using TOPFlash reporter assays and immunofluorescence for β-catenin localization
Designing comprehensive studies to evaluate CHST11 as a therapeutic target requires a multi-faceted approach:
Expression Analysis Across Cancer Types:
Patient Stratification Studies:
Classify patients based on CHST11 expression levels (high vs. low)
Correlate with clinical parameters (TNM staging, BCLC staging for liver cancer)
Perform survival analysis stratified by CHST11 expression and immune cell infiltration
Particularly focus on Treg-enriched vs. Treg-decreased patient subgroups
Mechanistic Studies:
| Approach | Methodology | Expected Outcome |
|---|---|---|
| Gene knockdown | siRNA or CRISPR-Cas9 | Changes in cell proliferation, metastasis, immune response |
| Gene overexpression | Stable transfection systems | EMT induction, increased migration, immune suppression |
| Pathway analysis | RNA-Seq after CHST11 modulation | Identification of key regulated pathways |
| Protein interaction | Co-IP, proximity labeling | Identification of CHST11 interaction partners |
Immune Microenvironment Assessment:
Preclinical Models:
Develop CHST11 inhibitors or antibody-drug conjugates targeting CHST11
Test in patient-derived xenografts and syngeneic mouse models
Evaluate combined blockade of CHST11 and immune checkpoints
Assess changes in tumor growth, metastasis, and immune infiltration
Translational Biomarker Development:
Establish immunoreactive score (IRS) cutoffs for CHST11 protein expression
Create and validate prognostic models incorporating CHST11 expression
Develop companion diagnostics to identify patients likely to benefit from CHST11-targeted therapy
Conduct receiver operating characteristic (ROC) curve analysis to assess diagnostic potential
This comprehensive approach will provide robust evidence for the potential of CHST11 as a therapeutic target, particularly for combination immunotherapy approaches.
Sample Preparation: Standard RIPA buffer with protease inhibitors
Protein Loading: 20-50 μg total protein per lane
Blocking: 5% non-fat milk or BSA in TBST, 1 hour at room temperature
Primary Antibody Incubation: Overnight at 4°C is optimal
Secondary Antibody: HRP-conjugated anti-rabbit or anti-mouse depending on primary antibody host
Special Considerations: Adding N-ethylmaleimide to lysis buffer can help preserve sulfation status
Antigen Retrieval: Citrate buffer (pH 6.0), high pressure
Blocking: 3% hydrogen peroxide followed by serum blocking
Primary Antibody Incubation: 1-2 hours at room temperature or overnight at 4°C
Detection System: HRP-polymer detection system
Counterstain: Hematoxylin
Analysis: Assess staining intensity and percentage in multiple fields (≥10) using 400× magnification
Scoring: Use immunoreactive scores (IRS) with cutoffs (e.g., IRS >6 for high expression)
Coating Buffer: 50 mM carbonate-bicarbonate, pH 9.6
Blocking: 1-5% BSA in PBS
Sample Types: Cell lysates, tissue homogenates, serum (depending on specific kit)
Detection: HRP-conjugated secondary antibody with TMB substrate
Sensitivity Enhancement: Avidin-biotin amplification if needed
Recommended Dilution: Start with 1:100-500
Fixation: 4% paraformaldehyde, 10-15 minutes
Permeabilization: 0.1-0.5% Triton X-100 in PBS, 5-10 minutes
Blocking: 5% normal serum from secondary antibody host species
Primary Antibody Incubation: Overnight at 4°C
Secondary Antibody: Fluorophore-conjugated (Alexa Fluor series recommended)
Counterstain: DAPI for nuclei visualization
Mounting: Anti-fade mounting medium
Cell Preparation: Single-cell suspension (1-5 × 10^6 cells/ml)
Fixation/Permeabilization: Required since CHST11 is intracellular
Recommended Dilution: 1:50-100
Blocking: 5% serum from secondary antibody host species
Primary Antibody Incubation: 30-60 minutes on ice
Secondary Detection: Fluorophore-conjugated secondary antibody
Controls: Include isotype controls and CHST11-knockdown cells
Potential Causes and Solutions:
Low CHST11 Expression: Confirm expression levels with RT-qPCR first
Insufficient Protein: Increase loading amount or concentrate samples
Antibody Concentration: Increase primary antibody concentration or incubation time
Protein Degradation: Add fresh protease inhibitors and keep samples cold
Transfer Issues: Optimize transfer conditions for higher molecular weight proteins
Detection Sensitivity: Use enhanced chemiluminescence or switch to more sensitive detection systems
Potential Causes and Solutions:
Non-specific Binding: Increase blocking time/concentration; try different blocking agents
Antibody Concentration: Dilute primary and secondary antibodies
Washing: Increase washing duration and number of washes
Secondary Antibody Cross-Reactivity: Use secondary antibodies pre-adsorbed against other species
Sample Contamination: Prepare fresh samples with protease inhibitors
Potential Causes and Solutions:
Fixation Issues: Standardize fixation time and conditions
Antigen Retrieval: Optimize antigen retrieval method and duration
Tissue Thickness: Ensure consistent section thickness (4-5 μm recommended)
Endogenous Peroxidase: Extend hydrogen peroxide blocking step
Staining Evaluation: Use established scoring systems (IRS as used by Xiong et al. )
Uneven Staining: Ensure even application of antibody and adequate incubation volume
Potential Causes and Solutions:
Post-transcriptional Regulation: Assess miRNA expression that might target CHST11
Protein Stability: Perform protein degradation assays (e.g., cycloheximide chase)
Antibody Specificity: Validate using CHST11 knockdown/overexpression controls
Technique Sensitivity: Use more sensitive detection methods like digital PCR for RNA and proximity ligation assay for protein
Potential Causes and Solutions:
Fixation Over-crosslinking: Reduce fixation time or switch to milder fixatives
Insufficient Permeabilization: Optimize Triton X-100 concentration and time
Primary Antibody Access: Increase incubation time or try different epitope-targeting antibodies
Signal Amplification: Use tyramide signal amplification or brighter fluorophores
Photobleaching: Use anti-fade mounting media and minimize exposure to light
Potential Causes and Solutions:
Inadequate Permeabilization: Test different permeabilization reagents/protocols
Cell Viability: Use viability dyes to exclude dead cells from analysis
Compensation: Proper compensation settings for multicolor experiments
Antibody Titration: Determine optimal antibody concentration with titration experiments
Control Samples: Include positive and negative controls (CHST11 knockdown cells)
When selecting a CHST11 antibody for research, several critical factors should be considered:
Epitope Specificity:
Identify which region of CHST11 the antibody targets
Different commercial antibodies target distinct regions:
Consider epitope conservation if working with non-human species
For functional studies, select antibodies targeting functional domains
Host Species and Clonality:
Validated Applications:
Species Reactivity:
Technical Specifications:
Experimental Validation Evidence:
Request validation data such as:
Western blot images showing single band at expected molecular weight (~42 kDa)
IHC images with positive and negative controls
Knockdown/overexpression validation
Peptide competition assays
Citation Record:
Batch Consistency:
For critical experiments, purchase larger amounts from single lots
For polyclonal antibodies, consider testing multiple lots side-by-side
For optimal results in CHST11 research, thorough antibody validation is recommended regardless of commercial claims, particularly comparing results with gene expression data and using genetic knockdown controls.
CHST11 antibodies provide powerful tools to investigate how this enzyme influences the tumor microenvironment:
Multiplex Immunofluorescence Profiling:
Use CHST11 antibodies in combination with markers for:
This approach allows visualization of spatial relationships between CHST11-expressing cells and immune cells
Quantify cell-to-cell distances and interactions using digital pathology platforms
Flow Cytometry for Immune Cell Phenotyping:
Combine CHST11 antibodies with immune cell markers in multiparameter flow cytometry
Sort CHST11-high versus CHST11-low tumor cells for further functional studies
Isolate tumor-infiltrating immune cells from these different microenvironments
Analyze expression of CHST11 within specific immune cell subsets
Tissue Microarray Analysis:
Co-culture Experimental Systems:
Establish co-cultures of CHST11-expressing cancer cells with:
Isolated T regulatory cells
PD-1/PD-L1-expressing immune cells
Use CHST11 antibodies to monitor expression during co-culture
Test effects of CHST11 blockade or knockdown on immune cell function
In Vivo Tumor Models with Imaging:
Develop fluorescently labeled CHST11 antibodies for in vivo imaging
Track CHST11 expression changes during tumor progression
Correlate with immune infiltration in real-time
Test therapeutic targeting of CHST11 in immunocompetent models
Mechanistic Studies:
Use CHST11 antibodies to immunoprecipitate the protein and identify binding partners
Perform ChIP assays to determine how CHST11 regulates genes involved in PD-L1/PD-1 pathways
Investigate post-translational modifications of CHST11 during tumor-immune interactions
Therapeutic Response Prediction:
Apply CHST11 IHC to pre-treatment biopsies from patients receiving immunotherapy
Correlate expression with clinical response
Develop CHST11-based companion diagnostic approaches for immunotherapy selection
These approaches can provide critical insights into how CHST11 contributes to immunosuppressive tumor microenvironments and may lead to novel combination immunotherapy strategies.
Based on current findings, several promising research directions for CHST11 emerge:
Combination Immunotherapy Development:
Targeting CHST11 may inhibit Treg infiltration and enhance the antineoplastic effect of immune checkpoint inhibitors
Design studies testing CHST11 inhibitors combined with PD-1/PD-L1 blockade
Develop novel Treg-modulating agents that target CHST11-dependent pathways
Establish optimal sequencing and dosing of combination approaches
Biomarker Development for Precision Medicine:
Validate CHST11 as a predictive biomarker for immunotherapy response
Develop standardized IHC protocols and scoring systems for clinical implementation
Create multi-marker panels including CHST11 and related immune markers
Design liquid biopsy approaches to monitor CHST11 expression non-invasively
Expanded Cancer Type Investigations:
While CHST11 has been studied in HCC and breast cancer, research should expand to:
Perform comparative analyses across cancer types to identify common mechanisms
Beyond Cancer: CHST11 in Other Diseases:
Investigate CHST11's role in autoimmune conditions given its impact on regulatory T cells
Explore connections to Osteochondrodysplasia and Brachydactyly (associated CHST11 diseases)
Study CHST11 in mucinoses and cartilage-related disorders
Examine potential roles in inflammatory diseases with dysregulated immune responses
Advanced Structural and Biochemical Studies:
Determine crystal structure of CHST11 to facilitate drug design
Characterize the specific sulfation patterns produced by CHST11 in different cellular contexts
Develop selective CHST11 inhibitors through structure-based drug design
Investigate the enzymatic interplay between CHST11 and other sulfotransferases
Novel Technological Approaches:
Single-cell analysis of CHST11 expression and its relationship to cellular phenotypes
Spatial transcriptomics to map CHST11 expression within complex tissue architectures
CRISPR screens to identify synthetic lethality partners for CHST11
Glycoproteomics to characterize CHST11-dependent changes in cellular glycosylation patterns
Developmental and Stem Cell Biology:
Examine CHST11's role in epithelial-mesenchymal transition during development and cancer
Investigate connections between CHST11, the Wnt pathway, and cancer stem cell properties
Study the regulation of CHST11 during cellular differentiation
Explore CHST11's potential as a target for cancer stem cell elimination
These research directions could significantly advance our understanding of CHST11 biology and lead to novel therapeutic strategies for cancer and other diseases.
Integrating CHST11 research with broader glycobiology can enhance our understanding of sulfation patterns and their biological significance:
Comprehensive Glycosaminoglycan (GAG) Analysis:
Since CHST11 catalyzes the 4-O-sulfation of chondroitin sulfate , researchers should:
Analyze complete sulfation patterns using mass spectrometry
Compare chondroitin sulfate structures in normal vs. CHST11-overexpressing tissues
Investigate how altered sulfation affects binding to growth factors and cytokines
Develop imaging mass spectrometry approaches to visualize sulfation patterns in situ
Integrative Analysis of Sulfotransferases:
CHST11 belongs to the sulfotransferase 2 family , suggesting integrated studies:
Compare expression patterns of multiple sulfotransferases across tissues and cancer types
Investigate functional redundancy and compensatory mechanisms
Study co-regulation of sulfotransferases in response to inflammatory signals
Examine interactions between different sulfotransferases in the Golgi apparatus
Proteoglycan Carrier Investigation:
Research shows CSPG4 (chondroitin sulfate proteoglycan 4) as a carrier for CHST11-mediated sulfation :
Characterize the full repertoire of proteoglycans modified by CHST11
Investigate how proteoglycan core proteins direct CHST11 activity
Study the trafficking and localization of CHST11 in relation to proteoglycan synthesis
Develop proteomics approaches to identify all CHST11-modified proteoglycans
Glycosylation-Immune System Connections:
CHST11's impact on immune infiltration suggests important glyco-immunology studies:
Investigate how sulfated GAGs bind to chemokines that attract regulatory T cells
Study how sulfation patterns affect antigen presentation and T cell recognition
Examine the impact of CHST11-dependent sulfation on immune checkpoint receptors
Develop glyco-engineered antibodies targeting CHST11-modified structures
Methodological Approaches for Integrated Studies:
| Technique | Application for CHST11 Research | Integration with Glycobiology |
|---|---|---|
| Glycomic profiling | Characterize sulfation patterns | Compare with other glycosylation changes |
| Lectin arrays | Detect changes in glycan recognition | Include sulfation-sensitive lectins |
| Glycoprotein enrichment | Isolate CHST11-modified proteins | Combine with proteomic identification |
| Metabolic labeling | Track synthesis of sulfated GAGs | Integrate with other glycan labeling |
| Glycan imaging | Visualize sulfated structures | Multimodal imaging of glycan types |
P-selectin Binding Studies:
Research has shown CHST11 expression correlates with P-selectin binding :
Investigate how CHST11-dependent sulfation affects all selectin interactions
Study the role of sulfated GAGs in cancer cell adhesion and metastasis
Develop inhibitors of sulfation-dependent selectin binding
Explore the relationship between CHST11, P-selectin, and platelets in metastasis
Translational Glycobiology Approaches:
Develop glycan-based biomarkers for cancer detection and monitoring
Design therapeutic approaches targeting specific sulfation patterns
Create antibodies recognizing CHST11-modified glycan epitopes
Investigate dietary or pharmacological modulators of sulfation pathways