HCST (Hematopoietic Cell Signal Transducer), also known as DAP10 or KAP10, is a transmembrane signaling adaptor protein encoded by the HCST gene in humans . It belongs to the DAP10 family and plays a critical role in immune recognition and signaling, particularly in natural killer (NK) and T cell responses . Synonyms include Phosphoinositide-3-Kinase Adaptor Protein, Transmembrane Adapter Protein KAP10, and Membrane Protein DAP10 .
HCST is integral to immunoreceptor complexes, such as NKG2D, which binds stress-induced ligands (e.g., MICA, MICB, ULBPs) . Its primary functions include:
Activation of NK and T Cells: Triggers cytotoxicity against MHC class I-associated ligands .
Cell Survival and Proliferation: Mediates PI3K/AKT and MAPK pathways to sustain immune cell viability .
HCST is significantly upregulated in KIRC tissues compared to normal kidney tissues, as shown by analyses of TCGA, GEO, and ULACAN datasets . Key findings:
| KEGG Pathway | Count | p-value | Immune/Cellular Role |
|---|---|---|---|
| Cell Adhesion Molecules (CAMs) | 29 | 4.71E-13 | Tumor-cell interaction and metastasis |
| Cytokine-Cytokine Receptor Interaction | 29 | 1.68E-07 | Immune signaling and inflammation |
| Natural Killer Cell Cytotoxicity | 23 | 1.03E-09 | Antitumor immunity |
| Antigen Processing and Presentation | 20 | 4.02E-11 | Immune recognition |
| Chemokine Signaling | 27 | 8.89E-09 | Leukocyte recruitment |
HCST interacts with immune-related proteins, including TYROBP, KLRC4, and PIK3R1, to modulate downstream pathways . Gene set enrichment analysis (GSEA) reveals HCST’s involvement in:
Immune Response: Antigen presentation, T cell receptor signaling, and Fcγ-mediated phagocytosis .
Tumorigenesis: PI3K/PTEN pathway dysregulation, proteasome activity, and cytosolic DNA sensing .
| Pathway | NOM p-value | Biological Impact |
|---|---|---|
| Proteasome | 0.005976096 | Protein degradation and tumor progression |
| Cytosolic DNA Sensing | 0.011292053 | Inflammation and immune activation |
| Hematopoietic Cell Lineage | 0.00501E-10 | Immune cell development |
HCST is detected via flow cytometry using monoclonal antibodies (e.g., Mouse Anti-Human DAP10/HCST Antibody, Catalog # MAB9786) . Intracellular staining protocols involve fixation and permeabilization with kits like FlowX FoxP3 Fixation & Permeabilization Buffer .
Hematopoietic Cell Signal Transducer (HCST) belongs to the DAP10 family and is involved in forming immunoreceptor complexes. It plays a crucial role in initiating cytotoxic responses against MHC class I chain-associated molecules like MICA and target cells displaying surface ligands such as UL16-binding protein (ULBP) and MICB. The HCST complex contributes to cell proliferation and survival by activating T cell and natural killer (NK) cell responses.
Produced in Sf9 insect cells using a baculovirus expression system, HCST is a single, glycosylated polypeptide chain consisting of 271 amino acids (specifically, residues 20 to 48). It has a molecular weight of 30.1 kDa. On SDS-PAGE analysis, the apparent molecular size ranges from 28 to 40 kDa.
A 242 amino acid human IgG-His tag is fused to the C-terminus of HCST. The protein is purified using proprietary chromatographic techniques.
The HCST protein solution is provided at a concentration of 1 mg/ml. It is formulated in a solution containing 10% glycerol and Phosphate Buffered Saline at a pH of 7.4.
The purity of HCST is greater than 90% as determined by SDS-PAGE analysis.
Hematopoietic Cell Signal Transducer, DNAX-Activation Protein 10, Phosphoinositide-3-Kinase Adaptor Protein, Transmembrane Adapter Protein KAP10, Kinase Assoc Pro Of ~10kDa, Membrane Protein DAP10, PIK3AP, DAP10, KAP10, Kinase Assoc Protein.
Sf9, Baculovirus cells.
ADPTTPGERS SLPAFYPGTS GSCSGCGSLS LPLEPKSCDK THTCPPCPAP ELLGGPSVFL FPPKPKDTLM ISRTPEVTCV VVDVSHEDPE VKFNWYVDGV EVHNAKTKPR EEQYNSTYRV VSVLTVLHQD WLNGKEYKCK VSNKALPAPI EKTISKAKGQ PREPQVYTLP PSRDELTKNQ VSLTCLVKGF YPSDIAVEWE SNGQPENNYK TTPPVLDSDG SFFLYSKLTV DKSRWQQGNV FSCSVMHEAL HNHYTQKSLS LSPGKHHHHH H.
HCST is a protein-coding gene involved in immune system function, particularly in signaling transduction pathways within immune cells. Gene Ontology (GO) annotations indicate that HCST and its co-expressed genes are significantly enriched in immune response regulation, T cell receptor signaling pathways, and intracellular signal transduction . HCST appears to function as a critical component in multiple immune-related processes including inflammatory responses and cell-cell signaling.
To investigate HCST's primary function, researchers should employ molecular techniques such as RNA sequencing to measure expression levels, co-immunoprecipitation to identify protein interactions, and functional assays to assess its role in immune cell activation. Gene Set Enrichment Analysis (GSEA) has demonstrated that HCST overexpression associates with antigen processing and presentation, cell adhesion molecules, and cytokine-cytokine receptor interactions . These techniques provide complementary approaches to characterize HCST's functional role in human biology.
HCST expression regulation appears to be tissue-specific and notably altered in disease states. Research data reveals significant differences in HCST expression between normal kidney tissue and kidney renal clear cell carcinoma (KIRC) tissue . To properly investigate these expression patterns, researchers should implement a multi-faceted approach including:
Comparative transcriptomics using RNA-seq data from matched normal and disease tissues
Epigenetic profiling including methylation analysis and histone modification assessment
Transcription factor binding studies using ChIP-seq to identify regulatory elements
Single-cell RNA sequencing to determine cell type-specific expression patterns
These methodologies allow researchers to elucidate the complex regulatory mechanisms governing HCST expression under both physiological and pathological conditions. Particularly valuable are data repositories such as TCGA and GEO, which contain expression profiles that can be analyzed using statistical approaches like t-tests to quantify differential expression between normal and diseased states .
According to GO annotation analysis, HCST and its co-expressed genes participate in multiple critical biological processes:
Signaling transduction
Inflammatory response
Apoptotic processes
Regulation of immune response
T cell receptor signaling pathway
Intracellular signal transduction
MHC class II protein complex interactions
Positive regulation of cell proliferation
KEGG pathway analysis further reveals that HCST co-expressed genes are significantly enriched in:
| Pathway | Count | P value |
|---|---|---|
| Cell adhesion molecules (CAMs) | 29 | 4.71E-13 |
| Cytokine-cytokine receptor interaction | 29 | 1.68E-07 |
| Chemokine signaling pathway | 27 | 8.89E-09 |
| Natural killer cell mediated cytotoxicity | 23 | 1.03E-09 |
| T cell receptor signaling pathway | 21 | 9.13E-10 |
| JAK-STAT signaling pathway | 15 | 0.001377994 |
| NF-kappa B signaling pathway | 14 | 2.54E-05 |
To investigate these processes experimentally, researchers should employ functional assays such as phosphorylation assays for signaling events, cytokine production assays for inflammatory responses, and co-culture systems to study cell-cell interactions in the context of HCST expression or inhibition.
Analysis from TCGA and GEO (GSE781 and GSE11151) databases demonstrates a significant relationship between HCST expression and KIRC. The data indicates that HCST expression is differentially regulated in KIRC tissues compared to normal kidney tissues . Methodologically, researchers investigating this relationship should:
Compare HCST mRNA and protein expression between matched normal and tumor samples
Correlate expression levels with clinicopathological features using multivariate statistical analyses
Perform immunohistochemistry to assess spatial expression patterns within tumor tissues
Evaluate expression in relation to tumor stage, grade, and other prognostic markers
HCST expression demonstrates significant correlation with cancer prognosis, particularly in KIRC. The following statistical approaches provide robust assessment of this relationship:
| Variable | HR | HR.95L | HR.95H | P value |
|---|---|---|---|---|
| HCST expression | 1.021210843 | 1.010143448 | 1.032399496 | 0.000159844 |
Multivariate analysis should be performed to determine if HCST is an independent prognostic factor when controlling for other variables like age, clinical stage, and tumor grade
Stratification analyses based on clinical factors should be conducted to identify subgroups where HCST expression has particularly strong prognostic significance
For methodological rigor, researchers should establish optimal expression cutoff values using approaches such as X-tile or receiver operating characteristic (ROC) curve analysis, and validate findings across independent cohorts. The significant association between HCST expression and clinical outcomes suggests its potential utility as a prognostic biomarker.
While the search results focus primarily on HCST's role in KIRC, KEGG pathway analysis reveals significant enrichment of HCST co-expressed genes in multiple immune-related disease pathways:
Autoimmune conditions:
Infectious diseases:
Inflammatory conditions:
To investigate HCST's role in these conditions, researchers should:
Perform comparative expression analyses across multiple disease types
Analyze patient samples to correlate HCST expression with disease severity
Develop animal models with altered HCST expression to assess disease susceptibility
Conduct in vitro functional studies with immune cells from patients with these conditions
The strong association of HCST with multiple immune-related pathways suggests broader implications beyond cancer, though direct causal relationships require further investigation through both bioinformatic approaches and experimental validation.
For comprehensive assessment of HCST expression, researchers should employ complementary techniques appropriate to their specific research questions:
Transcriptomic Analysis:
Quantitative RT-PCR: Provides high sensitivity for targeted expression analysis
RNA-Sequencing: Enables genome-wide expression profiling and detection of splice variants
Microarray: Useful for high-throughput screening across multiple samples
Protein-Level Analysis:
Western Blotting: Quantifies total protein expression and can detect post-translational modifications
Immunohistochemistry: Visualizes spatial expression patterns in tissue context
Flow Cytometry: Ideal for analyzing expression in specific cell populations
Single-Cell Approaches:
Single-cell RNA-seq: Reveals cell-type specific expression patterns
CyTOF: Allows simultaneous assessment of multiple proteins at single-cell resolution
Imaging Mass Cytometry: Combines spatial resolution with high-parameter analysis
Functional Assessment:
Reporter Gene Assays: Measures transcriptional regulation of HCST
CRISPR-based Screening: Identifies regulators of HCST expression
For proper analysis of HCST expression data, researchers should employ appropriate statistical methods such as t-tests for comparing expression between groups, as demonstrated in the TCGA and GEO analyses . When analyzing expression in relation to clinical outcomes, Kaplan-Meier and Cox regression analyses provide robust assessment of prognostic significance .
Analysis of HCST co-expression networks requires a systematic approach combining bioinformatic and experimental validation methods:
Correlation Analysis:
Functional Enrichment Analysis:
Protein-Protein Interaction Network Construction:
Experimental Validation:
Co-immunoprecipitation to confirm direct protein interactions
Proximity ligation assays to visualize protein interactions in situ
CRISPR-based perturbation of network components to assess functional relationships
This multi-faceted approach enables researchers to move beyond simple expression analysis to understand the broader functional network in which HCST operates, providing insights into potential mechanistic roles and therapeutic targets. For visual representation, interaction networks should be displayed using tools such as Cytoscape with appropriate layout algorithms to highlight key relationships.
When investigating HCST in large genomic datasets, researchers should implement several advanced bioinformatic approaches:
Multi-Omics Integration:
Correlate HCST expression with genomic alterations (mutations, CNVs)
Integrate transcriptomic, proteomic, and epigenomic data
Apply methods such as similarity network fusion or multi-factor analysis
Machine Learning Algorithms:
Develop predictive models for HCST-related outcomes
Use feature selection algorithms to identify key variables associated with HCST expression
Implement supervised (random forest, SVM) and unsupervised (clustering) approaches
Network-Based Analysis:
Construct gene regulatory networks to identify master regulators of HCST
Apply weighted gene co-expression network analysis (WGCNA)
Identify network modules associated with specific phenotypes
Pathway Topology Analysis:
Consider directionality and interaction types in pathway analysis
Apply methods such as SPIA (Signaling Pathway Impact Analysis)
Identify rate-limiting steps in HCST-related pathways
Single-Cell Data Analysis:
Characterize cell-type specific expression patterns
Identify cellular populations with co-expression of HCST and interacting genes
Reconstruct developmental trajectories related to HCST expression
The research on HCST in KIRC demonstrates the value of integrated approaches, combining expression analysis across multiple databases (TCGA, GEO, GEPIA, UALCAN) with pathway enrichment and survival analyses . For clinical applications, these bioinformatic findings should be validated in independent cohorts using appropriate statistical methods and adjusted for multiple testing to minimize false positives.
HCST function intersects with multiple signaling pathways, as revealed by KEGG pathway analysis of HCST co-expressed genes:
| Pathway | P value |
|---|---|
| T cell receptor signaling pathway | 9.13E-10 |
| JAK-STAT signaling pathway | 0.001377994 |
| NF-kappa B signaling pathway | 2.54E-05 |
| Chemokine signaling pathway | 8.89E-09 |
| Natural killer cell mediated cytotoxicity | 1.03E-09 |
| Fc gamma R-mediated phagocytosis | 3.55E-04 |
| B cell receptor signaling pathway | 0.004867075 |
| Toll-like receptor signaling pathway | 0.021063179 |
GSEA analysis further confirmed significant enrichment of these pathways in the HCST overexpression group . To experimentally investigate these pathways, researchers should employ:
Phospho-specific Western blotting and flow cytometry to detect activation of pathway components following HCST modulation
Small molecule inhibitors of specific pathway nodes to assess dependency relationships
CRISPR-Cas9 gene editing to knockout HCST and evaluate effects on pathway activation
Reporter assays using constructs with pathway-specific response elements
Co-immunoprecipitation studies to identify direct interactions between HCST and pathway components
The diverse array of signaling pathways associated with HCST suggests it functions as a critical node in immune cell signaling networks. Pathway crosstalk should be carefully considered, as JAK-STAT, NF-κB, and T cell receptor signaling frequently interact in immune contexts. Researchers should design experiments that can distinguish direct versus indirect effects of HCST on these pathways.
HCST appears to significantly influence immune cell function in the tumor microenvironment, as evidenced by the enrichment of immune-related pathways among its co-expressed genes. To properly investigate this relationship, researchers should:
Employ multiplex immunohistochemistry or CyTOF to characterize:
HCST expression across immune cell subsets in the tumor microenvironment
Correlation between HCST expression and markers of immune activation/exhaustion
Spatial relationships between HCST-expressing cells and other components of the tumor ecosystem
Conduct functional studies including:
Ex vivo tumor-infiltrating lymphocyte assays with HCST modulation
Co-culture systems with tumor cells and immune cells under varying HCST conditions
In vivo models with conditional HCST knockout in specific immune populations
Analyze gene signature correlations:
Compare HCST expression with established immune infiltration signatures
Assess relationship to interferon response genes
Correlate with markers of immunosuppression
The strong association of HCST with T cell receptor signaling (P=9.13E-10) and natural killer cell-mediated cytotoxicity (P=1.03E-09) suggests it may directly modulate anti-tumor immune responses. The involvement in pathways related to antigen presentation, cytokine signaling, and cell adhesion molecules further indicates HCST may influence immune cell recruitment, activation, and effector functions within the tumor microenvironment.
HCST appears to have significant roles in both T cell receptor signaling and natural killer (NK) cell function based on pathway enrichment analyses:
T Cell Receptor Signaling:
Natural Killer Cell Function:
To experimentally investigate these roles, researchers should:
Perform HCST knockdown/overexpression in isolated T cells and NK cells
Measure TCR signaling events (ZAP70/LAT phosphorylation, calcium flux, ERK activation)
Assess NK cell degranulation, cytokine production, and cytotoxicity against target cells
Evaluate receptor clustering and immune synapse formation using high-resolution microscopy
Conduct phosphoproteomics to identify HCST-dependent signaling events
The involvement of HCST in both adaptive (T cell) and innate (NK cell) immune pathways suggests it may function as an integrative regulator of anti-tumor immunity. Mechanistic understanding of how HCST influences these cell types could inform the development of immunotherapeutic approaches targeting this axis.
HCST shows promise as a prognostic biomarker in cancer, particularly in KIRC. To utilize HCST expression as a biomarker, researchers should follow this methodological framework:
For optimal clinical translation, researchers should also investigate whether HCST expression can be reliably measured in liquid biopsies, evaluate its performance in comparison to established biomarkers, and determine if it provides additional prognostic information when combined with other markers in composite models.
The extensive pathway associations of HCST suggest several potential therapeutic strategies:
Direct HCST Targeting:
Develop neutralizing antibodies against HCST
Design small molecule inhibitors that disrupt HCST interactions
Employ RNA interference approaches in therapeutic contexts
Methodology should include target validation using in vitro and in vivo models
Pathway-Based Interventions:
Immunotherapeutic Approaches:
Given HCST's enrichment in immune pathways, combine HCST targeting with:
Immune checkpoint inhibitors
Adoptive cell therapies (CAR-T, CAR-NK)
Cancer vaccines
Assess synergistic potential through preclinical combination studies
Precision Medicine Strategy:
Identify patient subgroups most likely to benefit from HCST-targeted therapy
Develop companion diagnostics to measure HCST expression or activity
Design biomarker-stratified clinical trials
Cell-Based Therapeutic Engineering:
Modify HCST expression in therapeutic T cells or NK cells
Enhance natural killer cell cytotoxicity by manipulating HCST signaling
Develop HCST-CAR constructs leveraging its signaling properties
The development of these therapeutic strategies requires systematic preclinical evaluation, including assessment of on-target effects, off-target toxicities, and potential resistance mechanisms. Given HCST's roles in normal immune function, particular attention should be paid to potential immunotoxicities of HCST-targeting approaches.
Given HCST's strong association with immune pathways, it may significantly influence immunotherapy response. To investigate this relationship, researchers should:
The significant enrichment of HCST in immune-related pathways provides strong biological rationale for its potential influence on immunotherapy response. Furthermore, its association with several viral pathways (Herpes simplex, Epstein-Barr virus, Influenza A) suggests potential relevance to virally-driven cancers and their immunotherapy treatment.
Despite the accumulating evidence regarding HCST's role in cancer and immune function, several critical knowledge gaps remain that warrant further investigation:
Mechanistic understanding of how HCST influences cancer progression remains incomplete, particularly regarding its direct versus indirect effects on tumor cells versus the tumor microenvironment. Studies employing cell type-specific knockout models would help elucidate these mechanisms.
The specific binding partners and signaling complexes formed by HCST in different cellular contexts require further characterization through techniques such as proximity labeling proteomics and structural biology approaches.
While HCST shows prognostic significance in KIRC , its relevance across other cancer types remains largely unexplored. Pan-cancer analyses using multi-omics approaches would clarify the breadth of HCST's oncological importance.
The potential of HCST as a therapeutic target has yet to be systematically evaluated through preclinical models. Developing tools for HCST modulation (antibodies, small molecules, genetic approaches) and testing them in relevant disease models represents a critical next step.
The exact mechanisms by which HCST influences T cell and NK cell functions remain to be fully elucidated, particularly in the context of anti-tumor immunity and response to immunotherapy. This requires detailed immunological studies at both molecular and cellular levels.
Addressing these knowledge gaps through rigorous scientific investigation will advance our understanding of HCST biology and potentially reveal new therapeutic opportunities for cancer and immune-related diseases.
Based on current knowledge and identified gaps, future HCST research should prioritize several key directions:
Comprehensive characterization of HCST expression across normal and disease tissues beyond KIRC, using techniques like spatial transcriptomics to understand tissue-specific expression patterns and cell-cell interactions involving HCST-expressing cells.
Detailed mechanistic studies to elucidate how HCST modulates signaling pathways, particularly focusing on the JAK-STAT (P=0.001377994), NF-κB (P=2.54E-05), and T cell receptor (P=9.13E-10) pathways identified in pathway analyses .
Investigation of HCST's role in the response to cancer immunotherapy, including retrospective and prospective analyses of HCST expression in patients receiving checkpoint inhibitors and other immunotherapeutic approaches.
Development and validation of HCST as a clinical biomarker, moving beyond association studies to prospective validation and incorporation into multi-biomarker predictive models for cancer prognosis and treatment response.
Therapeutic targeting studies, beginning with proof-of-concept experiments in preclinical models and advancing to rational drug design approaches targeting HCST or its key interaction partners.
Exploration of HCST's roles in autoimmune and infectious diseases, building on the pathway enrichment findings that suggest involvement in these conditions .
Single-cell multi-omics approaches to understand HCST's function at unprecedented resolution, identifying cell populations where HCST plays particularly critical roles and characterizing its dynamic regulation during immune responses.
The Hematopoietic Cell Signal Transducer (HCST), also known as DNAX-activation protein 10 (DAP10), is a transmembrane signaling adaptor protein. It plays a crucial role in the immune system by participating in the activation of natural killer (NK) cells and certain subsets of T cells. This protein is encoded by the HCST gene in humans.
The HCST gene is located on chromosome 19 and encodes a protein that contains a YxxM motif in its cytoplasmic domain . This motif is essential for the activation of phosphatidylinositol 3-kinase (PI3K) dependent signaling pathways . The protein is a part of the DAP10 family and is capable of forming an immunoreceptor complex .
HCST is primarily involved in the immune recognition receptor complex with the C-type lectin-like receptor NKG2D . This receptor complex is crucial for the activation of NK and T cell responses. The ligands for this receptor include MHC class I chain-related proteins (MICA and MICB) and UL16-binding proteins (ULBPs), which are upregulated under stress conditions such as viral infections and tumor transformations .
When the NKG2D receptor binds to its ligands, HCST activates PI3K signaling pathways through its YxxM motif . This activation leads to the induction of cytotoxicity against target cells expressing these ligands . The HCST-NKG2D receptor complex plays a significant role in cell survival and proliferation by triggering cytotoxic responses .
Mutations or dysregulation of the HCST gene can be associated with various diseases. For instance, HCST has been linked to conditions such as intracranial berry aneurysms and peroxisome biogenesis disorders . Understanding the function and regulation of HCST is essential for developing therapeutic strategies targeting immune responses in diseases like cancer and viral infections.
Recombinant HCST is produced using genetic engineering techniques to express the HCST protein in a host system, such as bacteria or mammalian cells. This recombinant protein is used in research to study its structure, function, and role in immune signaling pathways. It is also utilized in developing therapeutic interventions aimed at modulating immune responses.