The MAP2K3 antibody (e.g., catalog number 80137-1-RR) is validated for use in:
Western Blot (WB): Detects MAP2K3 in lysates from glioma cells (e.g., SH-SY5Y), hepatocellular carcinoma cells (HepG2), and fibrosarcoma cells (HT-1080) .
Immunofluorescence (IF)/Immunocytochemistry (ICC): Visualizes MAP2K3 localization in cytoplasmic regions of HeLa cells .
ELISA: Utilized for quantifying MAP2K3 protein levels in lysates or serum samples .
| Application | Dilution Range |
|---|---|
| Western Blot | 1:2000–1:10,000 |
| Immunofluorescence | 1:200–1:800 |
MAP2K3 expression correlates with glioma malignancy and poor prognosis. Immunohistochemistry using anti-MAP2K3 antibodies (e.g., HPA043783) revealed:
Higher protein levels in high-grade gliomas (HGG) compared to low-grade gliomas (LGG) and normal brain tissue .
Mutant p53 (e.g., R273H, R175H) upregulates MAP2K3 transcription via NF-Y and NF-κB cofactors . Studies using MAP2K3 antibodies demonstrated:
Increased protein levels in human tumor cells with mutant p53 .
MAP2K3 knockdown inhibits proliferation and survival in mutant p53-expressing cancer cells .
MAP2K3 inhibitors show promise in colorectal cancer models by selectively killing tumor cells (PMID: 31695024, 30770795) . The antibody is critical for validating MAP2K3 expression in preclinical studies.
| Sample Type | Positive Detection |
|---|---|
| Tumor Tissues | Glioma, hepatoma |
| Cell Lines | HeLa, HepG2, HT-1080 |
| Zebrafish Tissue | Embryonic extracts |
The role of MAP2K3 in various cellular processes has been extensively studied. Here are some key findings:
MAP2K3 is a member of the dual specificity kinase group that plays a key role in cell differentiation, motility, division, and death. It primarily functions by phosphorylating and activating the p38 MAPK signaling pathway in response to cytokines and environmental stress stimuli. MAP2K3 can be phosphorylated at sites Ser189 and Thr193 by MKKK proteins (MEKK 1-4) . Recent research highlights MAP2K3's potential as a therapeutic target, particularly in colorectal cancer where targeting MAP2K3 may represent an effective strategy to selectively kill cancer cells .
Commercial MAP2K3 antibodies support various applications, with Western Blot (WB) being the most validated. For instance, the 13898-1-AP antibody has been extensively validated for Western Blot with recommended dilutions of 1:1000-1:4000. This antibody shows reactivity with human, mouse, and rat samples, making it suitable for comparative studies across these species . Other applications may include ELISA, immunohistochemistry, and immunofluorescence depending on the specific antibody formulation and validation status.
When selecting a MAP2K3 antibody, researchers should consider:
Experimental application: Ensure the antibody is validated for your specific application (WB, IHC, IF, etc.)
Species reactivity: Verify compatibility with your experimental model (human, mouse, rat)
Antibody type: Consider whether polyclonal or monoclonal antibodies better suit your research needs
Epitope recognition: Determine if you need an antibody that recognizes specific phosphorylated forms or total MAP2K3
Molecular weight detection: Confirm the antibody detects the expected molecular weight (MAP2K3 is typically observed at 36-40 kDa)
For optimal preservation of antibody activity, MAP2K3 antibodies should be stored at -20°C in appropriate buffer conditions. For example, the 13898-1-AP antibody is stable for one year after shipment when stored at -20°C in PBS with 0.02% sodium azide and 50% glycerol (pH 7.3) . Importantly, for this particular formulation, aliquoting is unnecessary for -20°C storage. Researchers should avoid repeated freeze-thaw cycles and follow manufacturer-specific guidelines, as storage conditions may vary between products.
MAP2K3 antibodies can be employed to elucidate the role of this kinase in cancer through several methodological approaches:
Expression analysis in tumor vs. normal tissue: Immunohistochemistry with anti-MAP2K3 antibodies can reveal differential expression patterns. Research has shown aberrantly high MAP2K3 expression in various tumor tissues, including gliomas, correlating with poor clinicopathological characteristics and outcomes .
Signaling pathway investigation: Western blot analysis using phospho-specific and total MAP2K3 antibodies can assess activation of the p38 MAPK pathway in response to various stimuli or therapeutic interventions.
Correlation with immune markers: MAP2K3 expression analysis combined with immune checkpoint markers can help understand the relationship between MAP2K3 and immune infiltration in tumors. Research has demonstrated a significant relationship between MAP2K3 expression and immunological checkpoints, immune-related genes, and immune infiltration in glioma .
Functional studies: Using MAP2K3 antibodies in conjunction with knockdown or overexpression systems enables researchers to study the functional consequences of altered MAP2K3 expression or activity.
When investigating MAP2K3 in relation to immune cell infiltration, researchers should consider:
Dual immunostaining approaches: Use MAP2K3 antibodies alongside immune cell markers to assess co-localization or expression patterns within the tumor microenvironment.
Computational analysis integration: Combine antibody-based detection methods with computational approaches such as CIBERSORT and ssGSEA algorithms to correlate MAP2K3 expression with immune cell infiltration patterns .
ESTIMATE scoring implementation: Apply the "ESTIMATE" R package to determine immune, stromal, and ESTIMATE scores based on gene expression profiling and correlate these with MAP2K3 expression levels .
Immune checkpoint correlation analysis: Compare expression of various immunological checkpoints between high and low MAP2K3-expressing groups using appropriate statistical tests like the Wilcoxon rank sum test .
Therapy response prediction: Calculate Tumor Immune Dysfunction and Exclusion (TIDE) scores for high and low MAP2K3 expression groups to predict sensitivity to immune checkpoint inhibitor therapy .
When encountering challenges with MAP2K3 detection in Western blot applications, consider:
Antibody dilution optimization: Titrate the antibody (e.g., 1:1000-1:4000 range) to determine optimal concentration for your specific sample type .
Sample preparation variables: Different cell/tissue types may require adjusted lysis conditions to effectively extract MAP2K3. The protein has been successfully detected in various samples including A431 cells, mouse skeletal muscle tissue, A549 cells, THP-1 cells, and rat skeletal muscle tissue .
Molecular weight verification: Confirm bands appear at the expected molecular weight range of 36-40 kDa (as observed) compared to the calculated 39 kDa .
Phosphorylation state considerations: When studying activated MAP2K3, ensure samples are properly preserved to maintain phosphorylation at sites Ser189 and Thr193 .
Positive controls inclusion: Include validated positive controls such as A431 or A549 cells that are known to express detectable levels of MAP2K3 .
Loading control selection: Choose appropriate loading controls that don't overlap with MAP2K3's molecular weight range (36-40 kDa) to avoid signal interference.
To investigate MAP2K3 promoter regulation, researchers can employ these methodological approaches:
Promoter cloning and reporter assays: The MAP2K3 gene 5′-regulatory region can be PCR-amplified from genomic DNA (e.g., a 1.0 kb fragment from positions −989 to −2 relative to the translational start site) and cloned into a luciferase reporter vector like pGL3-Luc .
PCR amplification protocols: For human MAP2K3 promoter amplification, researchers can use primers such as:
ChIP assays: Chromatin immunoprecipitation can identify transcription factors binding to the MAP2K3 promoter. Quantitative PCR can be performed using primers specific to the hMAP2K3 promoter:
Expression verification: After promoter studies, cellular MAP2K3 expression can be verified by Western blot using appropriate antibodies to confirm the functional consequences of promoter regulation .
MAP2K3 antibodies can be instrumental in characterizing cancer-associated MAP2K3 mutants through:
When investigating post-translational modifications of MAP2K3, researchers should consider:
Phospho-specific antibody selection: For MAP2K3 activation studies, use antibodies that specifically recognize phosphorylated Ser189 and Thr193 sites, which are critical for its activation by upstream MKKK proteins .
Sample preparation optimization: Phosphorylation states are labile, so samples must be collected and processed with phosphatase inhibitors to preserve modification status.
Positive control stimulation: Include samples treated with known activators of the p38 MAPK pathway, such as cytokines or environmental stress inducers, as positive controls for MAP2K3 phosphorylation .
Kinase assay integration: Complement antibody-based detection with in vitro kinase assays to directly assess MAP2K3 enzymatic activity toward p38 MAPK substrates.
Mass spectrometry validation: For comprehensive phosphorylation site mapping, combine immunological methods with mass spectrometry analysis.
To study MAP2K3's role in immune regulation, researchers can design experiments that:
Utilize gene expression manipulation:
Assess immune infiltration consequences:
Determine clinical implications:
Implement pathway analysis:
To ensure antibody specificity in MAP2K3 research, consider:
Knockout/knockdown controls:
Multiple antibody validation:
Compare results using antibodies targeting different epitopes of MAP2K3
Verify consistent results across different antibody clones or sources
Peptide competition assays:
Pre-incubate the antibody with excess MAP2K3 immunizing peptide
Demonstrate signal reduction/elimination in the presence of competing peptide
Cross-reactivity assessment:
Molecular weight verification:
MAP2K3 antibodies offer valuable tools for translational cancer research through:
Tissue microarray analysis:
Prognostic biomarker validation:
Predictive biomarker development:
Therapeutic target evaluation:
For analyzing MAP2K3 in clinical samples, researchers should consider:
Immunohistochemistry protocol optimization:
Standardize antigen retrieval methods (heat-induced epitope retrieval is often effective)
Determine optimal antibody dilution through titration experiments
Include appropriate positive and negative control tissues in each run
Use The Human Protein Atlas database for reference images of normal brain tissue, LGG, and HGG samples
Sample preservation considerations:
Formalin-fixed paraffin-embedded (FFPE) versus fresh-frozen tissue processing
Immediate fixation to preserve protein phosphorylation status
Standardized fixation times to ensure consistent results
Quantification methods:
Implement digital pathology tools for objective quantification
Establish scoring criteria for staining intensity and percentage of positive cells
Validate scoring through multiple independent observers
Multi-omic integration: