STK4 forms a transcriptional complex with Foxp3 and NF-κB p65 in regulatory T (Treg) cells, stabilizing their function. Loss of STK4 in Treg cells leads to autoimmune disorders due to impaired NF-κB signaling and Treg activation .
In STK4-deficient patients, abnormal T cell subsets, NK cells, and dendritic cells correlate with recurrent infections (e.g., Epstein-Barr virus) and dysregulated cytokine responses .
STK4 as a Tumor Suppressor: Low STK4 expression in clear cell renal cell carcinoma (ccRCC) correlates with poor prognosis and altered immune infiltration (e.g., reduced plasmacytoid dendritic cells and NK cells) .
Mechanistic Insights: STK4 regulates apoptosis and cell cycle progression, with deficiencies linked to malignancies like B cell lymphoma .
STK4 phosphorylates Foxp3 at serine-418, enhancing its stability and transcriptional activity in Treg cells .
Caspase-cleaved STK4 fragments phosphorylate histone H2B, potentially driving chromatin condensation during apoptosis .
Data from bulk RNA sequencing highlights STK4’s role in immune modulation:
| Immune Cell Type | Correlation with STK4 Expression | p-value |
|---|---|---|
| Plasmacytoid dendritic cells | Negative (r = -0.23) | < 0.001 |
| NK CD56 bright cells | Negative (r = -0.12) | 0.017 |
| T helper cells | Positive (r = 0.18) | < 0.001 |
STK4-associated pathways in ccRCC include:
Western Blot: Detects STK4 in Jurkat T cells, SH-SY5Y neuroblastoma cells, and glioblastoma lines .
Immunofluorescence: Localizes STK4 to the cytoplasm in SH-SY5Y cells .
Clinical Relevance: Used to study STK4 mutations in patients with lymphoproliferative disorders and recurrent infections .
STK4 (also known as MST1) is a cytoplasmic kinase structurally similar to the yeast Ste20p kinase that acts upstream of stress-induced mitogen-activated protein kinase cascades. The protein can phosphorylate myelin basic protein and undergoes autophosphorylation. STK4 is particularly important in research because it plays critical roles in:
Apoptosis regulation, where caspase-cleaved fragments of STK4 can phosphorylate histone H2B, correlating with chromatin condensation
Immune cell function, particularly T cell survival and proliferation
Tumor suppression, especially in contexts like hepatocellular carcinoma
STK4 deficiency is associated with autosomal recessive combined immunodeficiency (CID), making it a crucial target for immunological research .
When selecting an STK4 antibody, researchers should consider the functional domains of the protein and the specific research questions being addressed:
N-terminal domain antibodies: Useful for detecting full-length STK4 protein, as demonstrated in studies of STK4 deficiency where monoclonal antibodies directed against the N-terminus successfully detected wild-type protein in controls but not in patients with certain mutations
Kinase domain recognition: Important for studies investigating STK4's enzymatic function
C-terminal SARAH domain: Critical for detecting dimerization capabilities, as this domain functions as a hydrophobic platform for homo- and heterodimerization
Notably, mutations truncating only the C-terminal SARAH domain can still produce detectable protein (albeit at reduced levels), whereas other mutations leading to complete protein loss require careful antibody selection for detection of any residual protein .
Validation of STK4 antibody specificity should include:
siRNA knockdown controls: As demonstrated in published studies, comparing western blot results between control cells and cells transfected with STK4-specific siRNA provides strong evidence of antibody specificity
Patient-derived cell analysis: In cases where STK4-deficient patient samples are available, these provide excellent negative controls, as seen in studies where STK4 protein was undetectable in patient T cells while intermediate levels were observed in heterozygous parents
Protein expression correlation: Verify that detected protein levels correlate with mRNA expression levels as measured by RT-qPCR or RNA-Seq, though note that post-transcriptional mechanisms may result in discrepancies (as observed in a patient with a SARAH domain mutation who showed elevated mRNA but reduced protein levels)
Multiple antibody comparison: Using antibodies targeting different epitopes of STK4 can confirm specificity and reveal domain-specific detection capabilities
Based on published research, the most effective methods for detecting STK4 expression in patient samples include:
Western blot analysis: The gold standard for quantifying STK4 protein expression, allowing detection of both full-length and truncated forms. This method effectively distinguished between complete absence of STK4 protein in some patients versus reduced levels in others
Quantitative RT-PCR: Essential for measuring STK4 gene expression, particularly useful when protein levels are too low for reliable detection by immunoblotting
RNA-Seq: Provides comprehensive transcriptomic analysis, allowing detection of STK4 transcript levels in context with other genes, facilitating pathway analysis
Flow cytometry: While less commonly reported for STK4 itself, flow cytometry is valuable for assessing downstream effects of STK4 deficiency on immune cell populations and surface markers like PD-1
It's important to correlate findings across multiple detection methods, as discrepancies between mRNA and protein levels have been observed in certain STK4 mutations .
To assess functional impacts of STK4 deficiency or mutation, researchers can employ the following antibody-dependent approaches:
Downstream target assessment: Immunoblotting for downstream targets like FoxO3a, which shows reduced levels in STK4-deficient cells, providing evidence of functional impairment of STK4 signaling pathways
Phosphorylation status analysis: Using phospho-specific antibodies to detect activation states of STK4-regulated pathways, such as p65 and STAT3 phosphorylation, which are inversely associated with STK4 expression in certain contexts
Cellular functional assays with antibody detection:
Co-immunoprecipitation studies: To assess STK4 protein interactions, particularly those involving the SARAH domain that mediates homodimerization and interactions with other proteins
These techniques have been successfully employed to characterize the functional consequences of different STK4 mutations, revealing that some mutations (like those affecting only the SARAH domain) can result in milder phenotypes than complete loss-of-function mutations .
When performing immunoblotting with STK4 antibodies, the following controls should be included:
Positive controls:
Negative controls:
siRNA knockdown samples (when possible)
Known STK4-deficient patient samples (if available)
Heterozygous controls:
Loading controls:
Specificity controls:
Competing peptide assays
Multiple antibodies targeting different epitopes of STK4
Research has shown that proper controls are critical for interpreting STK4 expression data, particularly in cases where mutations result in truncated but partially functional protein rather than complete absence .
STK4 antibodies can be instrumental in exploring the interplay between STK4 and Toll-like receptor (TLR) signaling:
Co-immunoprecipitation studies: Using STK4 antibodies to pull down protein complexes can reveal interactions with TLR pathway components. Research has identified relationships between STK4 and TLR pathway mediators, particularly IRAK1
Comparative expression analysis: Immunoblotting for STK4 alongside TLR pathway components (like IRAK1) in paired samples can reveal inverse relationships, as observed in macrophages isolated from hepatocellular carcinoma patients where STK4 expression was inversely associated with IRAK1, IL-6, and phosphorylated p65 or STAT3
Pathway intervention studies: Combining STK4 antibody detection with TLR pathway inhibitors (such as IRAK1/4 inhibitors) can help determine causality in observed correlations. In mouse models, IRAK1/4 inhibitor treatment affected IL-6 levels and tumor development in STK4-deficient contexts
Stimulus-response experiments: Using STK4 antibodies to track protein levels and modifications following TLR stimulation (e.g., with LPS or bacterial components) can elucidate dynamic relationships between these pathways
These approaches can provide insights into how STK4 regulates inflammatory responses through TLR pathways, with implications for both infectious disease and cancer research.
Detecting truncated or mutant forms of STK4 presents several technical challenges that researchers should consider:
Epitope availability: Depending on the mutation, epitopes recognized by particular antibodies may be absent or altered. For example, C-terminal mutations may not affect detection by N-terminal-targeting antibodies, as seen in a patient with a mutation truncating only the SARAH domain where the protein was still detectable, albeit at reduced levels
Protein stability differences: Mutations can affect protein stability, leading to lower steady-state levels despite normal or even elevated mRNA expression. This was observed in a case where STK4 mRNA was significantly higher than in controls, but the protein level was significantly lower due to a C-terminal truncation affecting the SARAH domain
Dimerization-dependent detection: Since wild-type STK4 predominantly exists as homodimers, mutations affecting dimerization (particularly in the SARAH domain) may alter antibody accessibility or protein conformation
Nuclear vs. cytoplasmic localization: STK4 can shuttle between cytoplasmic and nuclear compartments depending on its activation state. Mutations affecting autophosphorylation or nuclear localization signals may alter this distribution, requiring subcellular fractionation for accurate detection
Cross-reactivity with paralogs: STK4 has an important paralog, STK3, which could potentially cross-react with some antibodies, requiring careful specificity testing
To address these challenges, researchers should consider using multiple antibodies targeting different regions of the protein and correlating protein detection with mRNA expression data.
STK4 antibodies play a crucial role in differentiating between clinical phenotypes of STK4 deficiency by enabling detailed molecular characterization:
Residual protein detection: Different mutations in STK4 can lead to varying levels of protein expression, from complete absence to reduced levels of full-length or truncated protein. Western blotting with appropriate antibodies can quantify these differences, which have been correlated with clinical severity
Functional domain analysis: Using domain-specific antibodies can reveal whether particular functional domains remain intact. For instance, a patient with a mutation affecting only the SARAH domain exhibited a milder clinical phenotype despite severe T cell lymphopenia, contrasting with more severe presentations in patients with complete protein loss
Downstream pathway assessment: Immunoblotting for STK4-regulated proteins (like FoxO3a) provides insight into the functional consequences of different mutations. The degree of dysregulation in these pathways correlates with clinical manifestations
| STK4 Mutation Type | Protein Detection | T Cell Phenotype | Clinical Severity | Representative Features |
|---|---|---|---|---|
| Complete loss of protein | Undetectable by Western blot | Severe lymphopenia with impaired proliferation and increased apoptosis | Severe | Recurrent infections, EBV-associated lymphoproliferation, autoimmunity |
| SARAH domain truncation | Reduced but detectable protein | Lymphopenia with less impaired proliferation | Milder | Controlled sinopulmonary infections, absence of EBV complications |
| Other partial function mutations | Variable detection | Variable | Variable | Varied presentation depending on residual function |
This molecular stratification using antibody-based techniques has important implications for treatment decisions, as patients with milder molecular phenotypes may require less aggressive interventions than those with complete STK4 deficiency .
STK4 antibodies are valuable tools for investigating STK4's tumor suppressor functions, particularly in liver cancer:
Expression correlation studies: Immunoblotting for STK4 in paired tumor and non-tumor tissues can reveal expression patterns associated with cancer progression. Research has shown that STK4 expression is markedly reduced in macrophages isolated from human hepatocellular carcinoma (HCC) patients
Pathway interaction analysis: Using STK4 antibodies alongside detection of inflammatory mediators (IL-6, phospho-STAT3, phospho-p65) can elucidate mechanisms by which STK4 regulates tumor-promoting inflammation. Studies have demonstrated inverse correlations between STK4 and these inflammatory markers in HCC patients
Serum biomarker evaluation: While direct antibody applications typically involve tissue or cellular analysis, research has shown that serum STK4 levels were specifically decreased in HCC patients with high levels of IL-6, suggesting potential for circulating STK4 as a diagnostic biomarker
Therapeutic intervention assessment: STK4 antibodies can help evaluate the effectiveness of targeted interventions in STK4-deficient contexts. For example, in STK4-deficient mice, treatment with an IRAK1/4 inhibitor reduced serum IL-6 levels and liver tumor numbers, suggesting mechanistic links between STK4 deficiency, inflammation, and tumorigenesis
These applications highlight STK4's potential dual role in both immune regulation and tumor suppression, with antibody-based detection methods providing crucial insights for translational research.
Researchers facing discrepancies between STK4 mRNA and protein levels should consider these methodological approaches:
Comprehensive protein stability assessment:
Pulse-chase experiments with protein synthesis inhibitors to determine protein half-life
Proteasome inhibitors (e.g., MG132) to assess involvement of proteasomal degradation
Analysis of post-translational modifications affecting stability
Translation efficiency evaluation:
Polysome profiling to assess mRNA translation status
Analysis of translation initiation factors interacting with STK4 mRNA
Targeted mutation impact studies:
For known mutations (like those affecting the SARAH domain), generate construct variants to directly test effects on protein stability
Complementation experiments in deficient cells to verify functional consequences
Structural biology approaches:
In silico modeling of how mutations affect protein folding and stability
Comparison with known effects of mutations in similar protein domains
These approaches are particularly relevant given published observations of a patient with a SARAH domain truncation mutation who exhibited significantly higher STK4 mRNA levels than controls but significantly lower protein levels, suggesting that the inability to homodimerize may have reduced protein stability .
STK4 antibodies provide powerful tools for exploring the complex interactions between STK4 signaling and immune response pathways:
Stimulus-response profiling: Using STK4 antibodies to track protein levels and modifications following immune stimulation can reveal regulatory dynamics. Studies have employed RNA-Seq and protein analysis of PBMCs stimulated with IFN-α/IFN-β or PMA/ionomycin to identify STK4-dependent gene expression patterns
Regulatory network mapping:
Combining STK4 immunoprecipitation with mass spectrometry can identify novel interaction partners in immune contexts
Chromatin immunoprecipitation (ChIP) with STK4 antibodies can reveal direct genomic targets
Single-cell analysis: Using STK4 antibodies in flow cytometry or mass cytometry (CyTOF) protocols allows correlation of STK4 expression with cellular phenotypes at single-cell resolution
Phospho-protein network analysis: Multiplex phospho-protein detection alongside STK4 can map signaling networks. Research has identified regulatory networks of IFN-α/IFN-β-responsive genes encompassing cytokine, chemokine, and adhesion factor genes that are indirectly regulated by STK4
| Stimulus | STK4-Regulated Pathways | Cellular Functions Affected | Detection Methods |
|---|---|---|---|
| IFN-α/IFN-β | Cytokine/chemokine expression, adhesion factor regulation | Cytotoxicity, cell death, lymphocyte adhesion and migration | RNA-Seq, RT-PCR, ELISA, Flow cytometry, Western blot |
| PMA/ionomycin | T cell activation pathways | T cell proliferation, apoptosis resistance | Flow cytometry, CFSE assays, Annexin V staining |
| Anti-Fas challenge | Apoptotic pathways | Activation-induced cell death | DAPI/Annexin V profiling |
These approaches have revealed that STK4 deficiency leads to marked dysregulation, but not complete abrogation, of IFN-regulated gene expression, highlighting the nuanced role of STK4 in immune signaling networks .
Researchers working with STK4 antibodies may encounter several technical challenges:
Low signal intensity: STK4 protein levels may be naturally low or reduced in certain conditions
Solution: Use enhanced chemiluminescence (ECL) substrates with higher sensitivity
Solution: Optimize protein extraction methods to ensure complete recovery
Solution: Increase antibody incubation time or concentration, following titration experiments
High background in Western blots:
Solution: Increase blocking time or concentration
Solution: Try alternative blocking agents (milk vs. BSA)
Solution: Optimize antibody dilution through systematic titration
Solution: Include additional washing steps with higher detergent concentration
Cross-reactivity with paralogs:
Solution: Verify antibody specificity using siRNA knockdown controls
Solution: Consider using monoclonal antibodies with validated epitope specificity
Solution: Include known STK4-deficient samples as negative controls when possible
Inconsistent results in patient samples:
Detection of truncated proteins:
Solution: Use antibodies targeting different epitopes
Solution: Optimize gel percentage and running conditions for lower molecular weight proteins
Solution: Consider using gradient gels to resolve proteins of varying sizes
These solutions are based on successful approaches documented in STK4 research, where careful optimization of detection methods revealed important biological insights about protein expression in various mutant contexts .
Optimizing STK4 antibody-based assays for different biological contexts requires specific adaptations:
Immune cells (PBMCs, T cells):
Liver tissues and hepatocytes:
Cell lines vs. primary cells:
Antibody titration: Primary cells often require different antibody concentrations than cell lines
Signal amplification: Primary cells may need more sensitive detection methods
Controls: Include relevant cell-type specific positive and negative controls
Tissue-specific considerations:
Fixation protocols: Optimize fixation methods to preserve epitope accessibility
Antigen retrieval: Different tissues may require specific retrieval methods
Autofluorescence management: Use appropriate quenching methods for immunofluorescence applications
These adaptations should be empirically determined for each experimental context, with careful documentation of optimization steps to ensure reproducibility across studies.
For accurate quantification of STK4 protein in comparative studies, researchers should follow these best practices:
Standardized loading controls:
Quantification methodology:
Statistical analysis:
Apply appropriate statistical tests (e.g., unpaired Student's t-test has been used to compare STK4 expression between patient and control samples)
Report both mean values and measures of variation (standard error of the mean or standard deviation)
Consider power calculations to determine appropriate sample sizes
Reporting standards:
Present both raw blot images and quantification graphs
Clearly state normalization methods
Report all relevant experimental details (antibody source, dilution, incubation conditions)
Validation with orthogonal methods:
Correlate protein levels with mRNA expression when possible
Consider using ELISA or other quantitative protein detection methods as complementary approaches
These practices have been implemented in published STK4 research, resulting in rigorous quantitative comparisons that revealed significant differences in protein expression between patient and control samples, even in cases with subtle phenotypic variations .