HK3 (Hexokinase 3) catalyzes the first step of glycolysis, converting glucose to glucose-6-phosphate. Unlike HK1 and HK2, HK3 exhibits lineage-specific expression in myeloid cells and leukocytes, with emerging roles in immune regulation and cancer biology .
HK3 Antibodies are immunoglobulin-based reagents designed to bind HK3 for applications like immunohistochemistry (IHC), Western blot (WB), and enzyme-linked immunosorbent assay (ELISA). These antibodies vary by host species, clonality, and reactivity profiles .
Human lung cancer tissue: Strong HK3 signal observed using Proteintech 13333-1-AP with TE buffer (pH 9.0) antigen retrieval .
Dilution range: 1:20–1:200 (13333-1-AP) vs. 1:500–1:2,000 (67803-1-Ig) .
Observed molecular weight: ~99 kDa in Raji cells and rat spleen .
Cross-reactivity: Commercial HK3 antibodies may show cross-reactivity with HK1/HK2 unless rigorously validated .
Storage: Most antibodies require -20°C storage with glycerol to prevent freeze-thaw damage .
Purification: Protein A/G affinity chromatography ensures high purity .
HK3 is upregulated during neutrophil/macrophage differentiation of CD34+ hematopoietic cells (13.9-fold mRNA increase in HL60 cells) .
CRISPR-edited HK3-null AML cells show reduced viability and increased apoptosis during differentiation, independent of glycolytic activity .
Correlation with immune scores: High HK3 expression associates with elevated stromal/immune scores in NSCLC (TCGA data) .
Immunotherapy prediction: Patients with high HK3 levels showed 333-fold better response to PD-1 inhibitors (Keytruda) .
Biomarker potential: HK3 expression inversely correlates with tumor purity and genomic instability in NSCLC .
Therapeutic targeting: HK3’s role in immune evasion highlights it as a candidate for combination therapies involving checkpoint inhibitors .
Hexokinase 3 (HK3) is an enzyme that phosphorylates glucose to produce glucose-6-phosphate (G6P), representing the first critical step in most glucose metabolism pathways . As a member of the hexokinase family, HK3 is allosterically inhibited by its product (G6P) . The importance of HK3 in research stems from its emerging roles in:
Correlation with immune infiltrates in tumor microenvironments
Prediction of immunotherapy responses, particularly in NSCLC patients
Metabolic regulation in specialized cell types, especially in immune and cancer cells
Recent research has revealed novel functions beyond glucose metabolism, including HK3-mediated O-GlcNAcylation of EP300, which plays a role in tumor immune evasion mechanisms . This finding suggests HK3 may serve as a promising therapeutic target for enhancing cancer immunotherapy efficacy.
Based on the search results, there are multiple types of HK3 antibodies available for research applications:
Each antibody offers different advantages depending on the research application. Monoclonal antibodies provide high specificity and reproducibility, while polyclonal antibodies may offer broader epitope recognition and potentially stronger signals in certain applications .
Analysis of The Cancer Genome Atlas (TCGA) data has revealed significant correlations between HK3 expression and both clinical and molecular characteristics in cancer, particularly in non-small cell lung cancer (NSCLC) .
Key findings include:
Cases with low HK3 expression tend to be more malignant entities with frequent genomic aberrations of driver oncogenes
HK3 expression is linked to immune responses and inflammatory activities based on gene ontology analysis
HK3 expression shows a significant trend in predicting efficacy of immunotherapy for patients receiving PD-1 inhibitor treatment (Keytruda)
These correlations suggest HK3 expression may serve as a potential biomarker for predicting immunotherapy response and understanding tumor immune microenvironments.
For optimal Western Blot results with HK3 antibodies, researchers should follow these evidence-based protocols:
Sample preparation:
Blocking and antibody incubation:
Detection and visualization:
Use appropriate HRP-conjugated secondary antibodies
Visualize with standard chemiluminescence detection systems
Expected results:
Note that optimization may be required depending on specific experimental conditions and sample types.
Based on the search results, the following protocol is recommended for IHC applications with HK3 antibodies:
Tissue preparation:
Antigen retrieval:
Antibody dilutions:
Detection system:
Validated positive controls:
Researchers should note that optimal dilutions may vary depending on tissue type and specific experimental conditions, so preliminary titration experiments are recommended.
To ensure the specificity and reliability of HK3 antibody detection, researchers should implement the following validation strategies:
Positive and negative control samples:
Knockdown/knockout validation:
Perform siRNA or CRISPR-mediated HK3 knockdown/knockout experiments
Compare antibody signal between wild-type and HK3-depleted samples
Orthogonal validation:
Multiple antibody validation:
Confirm results using different HK3 antibodies targeting distinct epitopes
Compare monoclonal (e.g., 67803-1-Ig) and polyclonal (e.g., 13333-1-AP) antibodies to exclude epitope-specific artifacts
Recombinant protein controls:
These validation approaches help ensure that experimental findings are not compromised by antibody cross-reactivity or non-specific binding.
Recent research has uncovered a novel mechanism connecting glycolysis and immune evasion in clear cell renal cell carcinoma (ccRCC) through HK3-mediated O-GlcNAcylation of EP300 .
Key findings include:
Mechanism of action:
HK3 maintains EP300 protein stability by regulating O-GlcNAcylation levels in ccRCC cells
O-GlcNAcylation of EP300 at Ser900 enhances its stability
Stabilized EP300 works with TFAP2A as a co-transcription factor to promote PD-L1 transcription
EP300 also functions as an acetyltransferase to stabilize PD-L1 protein
Immunological consequences:
Increased PD-L1 expression contributes to tumor immune evasion by inhibiting T-cell cytotoxicity
Inhibition of HK3 leads to reduced PD-L1 expression, which restores T-cell cytotoxicity both in vitro and in immunocompetent mice
Microenvironment interactions:
ccRCC exhibits interactive dynamics with tumor-associated macrophages (TAMs)
UDP-GlcNAc serves as a critical substrate for O-GlcNAcylation and facilitates TAMs polarization
HK3 expression in ccRCC cells is influenced by IL-10 secreted by M2 TAMs
This newly identified mechanism suggests targeting HK3 could be a promising strategy for overcoming immune evasion in cancer, potentially enhancing immunotherapy efficacy by restoring T-cell functions through downregulation of PD-L1.
The relationship between HK3 expression and macrophage polarization represents a complex bidirectional interaction within the tumor microenvironment, as evidenced by recent research .
Key aspects of this relationship include:
Influence of macrophages on HK3 expression:
HK3's role in macrophage polarization:
Markers of macrophage polarization:
Clinical relevance:
Potential therapeutic implications:
Targeting HK3 could potentially repolarize macrophages from M2 to M1 phenotype
This repolarization could enhance anti-tumor immune responses and potentially improve immunotherapy outcomes
This relationship highlights how metabolic enzymes like HK3 extend beyond their classical roles in energy metabolism to influence immune cell function and phenotype in the tumor microenvironment.
Analysis of clinical data has revealed significant correlations between HK3 expression and response to PD-1 inhibitor therapy, particularly with pembrolizumab (Keytruda) :
Predictive value:
Molecular mechanisms:
HK3-mediated O-GlcNAcylation of EP300 promotes PD-L1 expression at both transcriptional and protein levels
This mechanism provides a direct link between HK3 activity and the PD-1/PD-L1 immune checkpoint pathway
Inhibition of HK3 leads to reduced PD-L1 expression, potentially enhancing T-cell cytotoxicity
Clinical implications:
Future research directions:
Validation in larger clinical cohorts is needed
Investigation of combination strategies targeting both HK3 and immune checkpoints
Exploration of HK3 as a therapeutic target to enhance immunotherapy efficacy
These findings highlight the potential of HK3 as both a predictive biomarker and therapeutic target in the context of cancer immunotherapy.
Researchers may encounter several challenges when using HK3 antibodies for immunohistochemistry. Here are common issues and their solutions:
High background staining:
Problem: Non-specific binding leading to high background
Solutions:
Weak or no signal:
Problem: Insufficient antigen detection
Solutions:
Variable staining across tissue sections:
Edge artifacts:
Problem: Stronger staining at tissue edges
Solutions:
Ensure adequate fixation of tissues
Apply hydrophobic barrier around tissue sections
Increase washing volume and ensure entire slide is submerged
Cross-reactivity with other hexokinase isoforms:
Problem: Potential false positive signal from HK1, HK2, or HK4
Solutions:
Use monoclonal antibodies with validated specificity for HK3
Perform parallel staining with antibodies against other hexokinase isoforms
Include HK3 knockout/knockdown controls when possible
When troubleshooting, methodically change one variable at a time and document all modifications to identify optimal conditions for specific experimental systems.
Detecting low-abundance HK3 in Western blot applications requires careful optimization. Based on the search results and best practices, here are strategies for enhancing HK3 detection:
Sample enrichment strategies:
Increase total protein loading (up to 50-60 μg per lane)
Use tissues with known higher HK3 expression (e.g., Raji cells, Ramos cells, RAW 264.7 cells) as positive controls
Consider subcellular fractionation to concentrate HK3 (primarily cytoplasmic)
Use immunoprecipitation to enrich HK3 before Western blot
Protein extraction optimization:
Transfer and detection optimization:
Use wet transfer methods for high molecular weight proteins like HK3 (99 kDa)
Extend transfer time or reduce voltage for more complete transfer
Use PVDF membranes instead of nitrocellulose for higher protein binding capacity
Apply enhanced chemiluminescence (ECL) substrate with extended exposure times
Consider using highly sensitive detection systems (e.g., femto-level ECL substrates)
Antibody optimization:
Tissue-specific considerations:
For lymphoid tissues: Use gentler lysis buffers to preserve protein integrity
For highly glycolytic tissues: Include additional wash steps to reduce background
For muscle or fibrous tissues: Extend homogenization time for complete lysis
These optimization strategies should be implemented systematically, changing one variable at a time to identify the most effective protocol for each specific tissue type and experimental context.
When investigating HK3-mediated O-GlcNAcylation, researchers should include several critical controls to ensure experimental validity and interpretability:
Positive and negative controls for HK3 expression:
O-GlcNAcylation detection controls:
Use anti-O-GlcNAc antibodies (e.g., Cell Signaling Technology #9875) as used in published studies
Include samples treated with O-GlcNAcase inhibitors (e.g., Thiamet G) as positive controls
Include samples treated with OGT inhibitors (e.g., OSMI-1) as negative controls
Use tissues/cells known to have high O-GlcNAcylation levels as reference standards
Substrate-specific controls for EP300 O-GlcNAcylation:
Metabolic controls:
Glucose deprivation to reduce UDP-GlcNAc availability
Glucosamine supplementation to increase UDP-GlcNAc pool
Alternative hexokinase inhibition/expression to determine HK3-specificity
Assessment of UDP-GlcNAc levels using metabolomics approaches
Downstream pathway controls:
Including these comprehensive controls will ensure that observed effects can be specifically attributed to HK3-mediated O-GlcNAcylation rather than to other confounding factors or non-specific effects.
HK3 antibodies offer valuable tools for investigating the metabolic reprogramming of immune cells in the tumor microenvironment through several advanced experimental approaches:
Multiplex immunofluorescence (mIF) assays:
Combine HK3 antibodies with immune cell markers (e.g., CD8 for T cells, CD68 for macrophages)
Use anti-O-GlcNAc antibodies in conjunction with HK3 to assess glycosylation patterns
Include metabolic markers (e.g., GLUT1, PKM2) to build comprehensive metabolic profiles
Analyze spatial relationships between HK3-expressing cells and immune cells in the tumor microenvironment
Flow cytometry applications:
Perform intracellular staining for HK3 in isolated tumor-infiltrating immune cells
Combine with surface markers for immune cell identification and activation status
Use fluorescent glucose analogs (e.g., 2-NBDG) to correlate HK3 expression with glucose uptake
Sort HK3-high versus HK3-low immune populations for functional assays
Single-cell analysis:
Integrate HK3 antibody staining with single-cell RNA sequencing
Perform cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) with HK3 antibodies
Correlate HK3 protein levels with metabolic gene expression signatures at single-cell resolution
Map metabolic heterogeneity across immune cell subpopulations
Ex vivo functional assays:
Isolate tumor-associated macrophages and measure HK3 expression in M1 versus M2 phenotypes
Assess how HK3 inhibition affects macrophage polarization and cytokine production
Evaluate T cell effector functions (cytokine production, cytotoxicity) in relation to HK3 expression
Study how glucose availability affects HK3 expression and function in isolated immune cells
In vivo imaging applications:
Develop fluorescently-labeled HK3 antibodies for intravital microscopy
Track HK3 expression dynamically during immune cell recruitment to tumors
Correlate with metabolic imaging techniques (e.g., hyperpolarized MRI with 13C-pyruvate)
These approaches leverage HK3 antibodies to provide insights into how metabolic reprogramming influences immune cell function in the tumor microenvironment, potentially identifying new targets for immunometabolic cancer therapies.
The correlations between HK3 expression and immunotherapy response suggest significant potential for biomarker development with several important implications:
Predictive biomarker potential:
Studies have shown HK3 expression correlates with response to PD-1 inhibitor therapy (Keytruda)
This correlation suggests HK3 could complement existing biomarkers like PD-L1 expression and tumor mutational burden
Integration into multi-parameter predictive models could enhance patient selection accuracy
Technical considerations for clinical implementation:
IHC-based detection using validated antibodies (e.g., 67803-1-Ig, 13333-1-AP) offers practical implementation in clinical pathology workflows
Standardized scoring systems would need to be developed (e.g., H-score, percentage positive cells)
Quality control measures including positive controls (e.g., human lung cancer tissue) would be essential
Biological rationale for predictive value:
Mechanistic link between HK3, O-GlcNAcylation of EP300, and PD-L1 expression provides biological plausibility
HK3's role in macrophage polarization suggests it may indicate broader immune landscape features beyond just PD-L1 expression
Correlation with genomic aberrations of driver oncogenes suggests potential integration with genomic biomarkers
Therapeutic targeting implications:
Current limitations and research needs:
Larger prospective clinical validation studies are needed
Standardization of detection methods across laboratories
Understanding the impact of intratumoral heterogeneity of HK3 expression
Determining optimal cutoff values for high versus low HK3 expression
Development of HK3 as a biomarker would require thorough analytical and clinical validation, but the existing mechanistic and correlative evidence provides a strong foundation for further investigation in this direction.