MAPK10 antibodies are recombinant monoclonal or polyclonal antibodies designed to bind specifically to MAPK10, a serine/threonine kinase involved in stress response, apoptosis, and immune regulation . The protein is encoded by the MAPK10 gene (Gene ID: 5602) and functions in the JNK signaling pathway, influencing cellular processes like proliferation and differentiation .
MAPK10 antibodies have been critical in identifying MAPK10’s role as a prognostic marker in HCC. Studies using TCGA data revealed:
Downregulation in HCC: MAPK10 expression is reduced in HCC tumors, correlating with poor patient survival (P < 0.001) .
Immune Microregulation: Low MAPK10 expression associates with reduced tumor-infiltrating lymphocytes (TILs) and stromal cells, fostering an immunosuppressive tumor microenvironment (TME) .
ICAM1 Regulation: MAPK10 modulates ICAM1 expression (P < 0.0001 in vitro), a protein critical for immune cell recruitment .
Pathway Enrichment: MAPK10-linked differentially expressed genes (DIGs) are enriched in chemokine signaling, T-cell activation, and cytokine interactions .
Western Blot: ProMab’s antibody detects MAPK10 at ~53 kDa in HEK293, Hela, and Jurkat cell lines .
ELISA: Proteintech’s antibody pair (84875-4-PBS capture, 84875-3-PBS detection) enables sensitive quantification in sandwich assays .
MAPK10 antibodies facilitate the development of immunotherapy strategies by:
Current antibodies are restricted to human and rodent samples, necessitating expansion to other species. Further studies are required to explore MAPK10’s isoform-specific roles and therapeutic targeting.
MAPK10 (also known as JNK3) is a serine/threonine-protein kinase implicated in diverse cellular processes, including neuronal proliferation, differentiation, migration, and apoptosis. Activation of the stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) pathway is triggered by extracellular stimuli such as proinflammatory cytokines or physical stress. Within this cascade, MAP2K4/MKK4 and MAP2K7/MKK7 phosphorylate and activate MAPK10/JNK3. Subsequently, MAPK10/JNK3 phosphorylates various transcription factors, predominantly AP-1 components like JUN and ATF2, thereby modulating AP-1 transcriptional activity. MAPK10 plays crucial regulatory roles in neuronal apoptotic signaling pathways. It phosphorylates the microtubule regulator STMN2 and is involved in regulating amyloid-beta precursor protein (APP) signaling during neuronal differentiation via APP phosphorylation. Furthermore, it contributes to neurite growth in spiral ganglion neurons and phosphorylates the CLOCK-ARNTL/BMAL1 heterodimer, influencing photic regulation of the circadian clock. Finally, MAPK10 phosphorylates JUND, a process inhibited by MEN1.
Research Highlights on MAPK10/JNK3:
MAPK10, also known as JNK3, is a member of the MAP kinase family that acts as an integration point for multiple biochemical signals. It is specifically expressed in a subset of neurons in the nervous system and is activated by threonine and tyrosine phosphorylation . MAPK10 is involved in various cellular processes including neuronal proliferation, differentiation, migration, and programmed cell death. Its importance in research stems from its role in stress-induced neuronal apoptosis, regulation of the amyloid-beta precursor protein/APP signaling during neuronal differentiation, and its potential tumor suppressor functions in multiple cancers .
MAPK10 (JNK3) antibodies are specifically designed to target the unique epitopes of MAPK10 that distinguish it from other MAPK family members. While MAPK10 shares structural similarities with other JNK isoforms (JNK1/MAPK8 and JNK2/MAPK9), MAPK10 antibodies are raised against unique regions to ensure specificity . The main differences include:
Tissue-specific detection: MAPK10 antibodies target proteins primarily expressed in neural tissues, heart, and specific tumor types, whereas antibodies against other MAPKs (like ERK1/2) target more ubiquitously expressed proteins .
Phosphorylation-state specificity: Some MAPK10 antibodies are designed to recognize specific phosphorylated residues (Thr and Tyr) that indicate its activation state .
Isoform specificity: MAPK10 has multiple splicing variants, and certain antibodies may be developed to target specific isoforms .
To properly validate MAPK10 antibodies for research applications, the following methodological approach is recommended:
Western blot analysis: Verify a single band of the expected molecular weight (approximately 53 kDa for human MAPK10) .
Immunohistochemistry (IHC) controls: Include both positive controls (tissues known to express MAPK10, such as brain tissue) and negative controls (tissues with knockout or knockdown of MAPK10) .
Cross-reactivity testing: Validate against recombinant MAPK8, MAPK9, and MAPK10 proteins to confirm specificity .
Gene silencing validation: Confirm antibody specificity using siRNA or shRNA targeting MAPK10, as demonstrated in studies of HCC cell lines (e.g., Huh7-shMAPK10) .
Quantitative PCR correlation: Verify that protein levels detected by the antibody correlate with mRNA expression levels of MAPK10 .
Optimizing MAPK10 antibodies for IHC in cancer tissues requires specific methodological considerations:
| Staining Intensity | Score | Percentage of Positive Staining | Score |
|---|---|---|---|
| Absent expression | 0 | 0-9% | 0 |
| Weak expression | 1 | 10-25% | 1 |
| Moderate expression | 2 | 26-50% | 2 |
| Strong expression | 3 | 51-100% | 3 |
Control selection: Include both tumor and adjacent non-tumor tissues, as MAPK10 expression is frequently downregulated in cancer compared to normal tissues .
Detection of MAPK10 phosphorylation requires specific methodological approaches:
Antibody selection: Use phospho-specific antibodies that recognize dual phosphorylation at Thr-221 and Tyr-223 sites, which indicate active MAPK10 .
Sample preparation: Quick sample lysis and processing is critical as phosphorylation states can rapidly change. Include phosphatase inhibitors in lysis buffers to preserve phosphorylation status .
Experimental controls:
Detection methods:
Quantification: Calculate the ratio of phosphorylated to total MAPK10 to determine activation levels rather than absolute values of phosphorylated protein .
When studying MAPK10 expression in disease models, the following controls are essential:
Tissue/cell type controls:
Expression validation controls:
qPCR validation: Verify protein expression changes with mRNA level measurements using validated primers (e.g., endogenous MAPK10 sense primer 5′-CTTCCCAGATTCCCTCTTCC-3′ and antisense primer 5′-GCTGGGTCATACCAGACGTT-3′) .
Multiple antibody validation: Use antibodies targeting different epitopes of MAPK10 to confirm expression patterns.
Genetic manipulation controls:
Methylation status controls: For epigenetic regulation studies, include methylation-specific PCR controls such as YccB1 cell line (breast carcinoma with hypermethylated MAPK10) .
Clinical sample controls: When analyzing patient samples, include:
MAPK10 antibodies can be employed in several methodological approaches to investigate the tumor microenvironment (TME) in hepatocellular carcinoma:
Multiplex immunohistochemistry/immunofluorescence:
Analysis of MAPK10-dependent immune signaling:
Investigate the relationship between MAPK10 and immunity-related genes, particularly the 495 differentially expressed immune-associated genes (DIGs) identified in HCC, where 482 genes are downregulated and 13 genes are upregulated in parallel with decreased MAPK10 expression .
Focus on key hub genes including SYK, CBL, VAV1, LCK, and CD3G, which respond to immunological costimulatory signaling mediated by ICAM1 .
Functional validation experiments:
Use MAPK10 antibodies in chromatin immunoprecipitation (ChIP) assays to identify MAPK10's direct targets in immune regulation.
Combine with RNA-seq data to correlate MAPK10 binding with gene expression changes in the TME.
Clinical correlation studies:
Stratify HCC patient samples based on MAPK10 expression levels and correlate with:
TIL abundance and composition
Markers of immune activation/suppression
Patient survival outcomes
Research has demonstrated that MAPK10 expression significantly correlates with survival prognosis in HCC patients, with potential implications for immunotherapy response prediction .
Studying MAPK10 phosphorylation dynamics in neuronal systems presents several methodological challenges:
Temporal resolution limitations:
Spatial heterogeneity in neuronal populations:
Isoform-specific detection:
Pathway crosstalk complications:
Balancing signal detection sensitivity with energy costs:
Recent research has highlighted the energetic costs of MAPK signaling cascades, with implications for understanding physiological phosphorylation dynamics.
Solution: Consider the trade-off between accuracy of information transmission and energetic cost when designing experiments to study MAPK10 phosphorylation dynamics .
Resolving contradictory findings regarding MAPK10's dual role requires specific methodological approaches:
Context-dependent analysis:
Epigenetic regulation investigation:
Combine MAPK10 protein detection with DNA methylation analysis using methylation-specific PCR (MSP).
Research has shown that MAPK10 is frequently downregulated in hepatocellular carcinoma through DNA methylation (detected in 58% of HCC cell lines and 66% of primary HCC tissues) .
Primer pairs for methylation detection: MAPK10m3/MAPK10m5 (methylated alleles) and MAPK10u3/MAPK10u5 (unmethylated alleles) .
Isoform-specific functional studies:
Utilize antibodies that can distinguish between MAPK10 isoforms to determine if different variants have opposing functions.
Design experiments to examine splice variant-specific effects on cell proliferation, apoptosis, and migration.
Pathway interaction network analysis:
Combined prognostic marker approach:
Optimizing western blot protocols for MAPK10 detection requires specific technical considerations:
Sample preparation:
Use RIPA or NP-40 based lysis buffers with protease and phosphatase inhibitors.
For phosphorylated MAPK10 detection, immediate sample processing is crucial to prevent dephosphorylation.
Protein loading and transfer:
Load 20-50 μg of total protein per lane for standard detection.
Use PVDF membranes (rather than nitrocellulose) for improved retention of phosphorylated proteins.
Consider wet transfer methods for larger MAPK10 isoforms.
Antibody selection and dilution:
Signal detection optimization:
For low abundance MAPK10 detection, enhanced chemiluminescence (ECL) substrates with extended signal duration are recommended.
Consider using signal enhancement systems for detecting minor MAPK10 isoforms.
Stripping and reprobing protocols:
When analyzing both total and phosphorylated MAPK10 on the same membrane, use mild stripping buffers to preserve epitopes.
Alternatively, use separate blots from the same samples run in parallel.
Accurately quantifying MAPK10 expression in heterogeneous samples requires specialized approaches:
Laser capture microdissection:
Cell sorting prior to analysis:
Single-cell analysis techniques:
Implement single-cell western blotting or mass cytometry for direct quantification of MAPK10 at the single-cell level.
These approaches avoid the averaging effect seen in bulk tissue analysis.
Normalization strategies:
Use cell-type specific markers as denominators when calculating relative MAPK10 expression.
For example, normalize neuronal MAPK10 to neuronal markers rather than housekeeping genes expressed in all cell types.
Combination of immunohistochemistry with digital image analysis:
Implement multiplexed immunohistochemistry with MAPK10 antibodies alongside cell-type markers.
Use digital pathology tools to quantify MAPK10 expression specifically in cells of interest.
Researchers should be aware of these common pitfalls when interpreting MAPK10 antibody data in clinical samples:
Failure to account for tumor heterogeneity:
Incomplete scoring systems:
Lack of standardized cutoff values:
Insufficient controls:
Overlooking post-translational modifications:
Correlation versus causation errors:
Emerging research indicates MAPK10's role in circadian rhythm regulation through its interaction with the CLOCK-BMAL1 heterodimer . Methodological approaches to study this relationship include:
Temporal expression analysis:
Use MAPK10 antibodies in time-course experiments to track MAPK10 expression and phosphorylation across circadian cycles.
Implement synchronized cell culture models with sampling every 4 hours over a 24-48 hour period.
Co-immunoprecipitation (Co-IP) protocols:
Use MAPK10 antibodies for Co-IP experiments to pull down CLOCK-BMAL1 complexes.
Validate interactions by reverse Co-IP using CLOCK or BMAL1 antibodies to pull down MAPK10.
Chromatin immunoprecipitation (ChIP) analysis:
Employ MAPK10 antibodies in ChIP experiments to identify genomic regions where MAPK10 interacts with circadian regulatory elements.
Combine with sequencing (ChIP-seq) to create genome-wide maps of MAPK10 binding sites related to circadian regulation.
Phosphorylation-specific detection:
Functional readout systems:
Combine MAPK10 antibody-based detection methods with reporter systems for circadian genes.
Correlate MAPK10 activity with oscillations in circadian gene expression.
This research direction has significant implications for understanding how cellular stress response pathways interface with circadian timing mechanisms .
Recent research has highlighted the trade-off between accuracy of information transmission and energetic cost in MAPK signaling cascades . To study MAPK10's role in this context:
Quantitative phosphorylation analysis:
Use MAPK10 antibodies in quantitative western blotting or ELISA to measure the stoichiometry of phosphorylation.
Implement methods to distinguish between mono- and dual-phosphorylated forms of MAPK10.
ATP consumption assays:
Combine MAPK10 activity measurements with assays that quantify ATP consumption.
Correlate signaling accuracy with energetic expenditure under different cellular conditions.
Pathway modeling approaches:
Develop computational models incorporating MAPK10 signaling dynamics and energetic parameters.
Use experimental data from MAPK10 antibody-based studies to validate and refine these models.
Futile cycle analysis:
Design experiments to measure the rate of MAPK10 phosphorylation-dephosphorylation cycling.
Employ pulse-chase approaches with phosphorylation state-specific antibodies to track phosphorylation dynamics.
Cellular fitness correlations:
This emerging research direction offers a novel framework for understanding the evolutionary design principles of MAPK10 signaling in terms of balancing information accuracy with energetic efficiency .
Integration of MAPK10 antibodies with systems biology approaches represents an emerging frontier in understanding complex disease networks:
Multi-omics data integration:
Combine MAPK10 antibody-based proteomics with transcriptomics, metabolomics, and epigenomics data.
This integration helps place MAPK10 within broader cellular networks and identify non-obvious relationships.
Protein-protein interaction (PPI) network analysis:
Pathway cross-talk mapping:
Dynamic network modeling:
Use time-course data from MAPK10 antibody-based experiments to inform mathematical models of signaling dynamics.
These models can predict emergent properties not obvious from static analyses.
Network perturbation analysis:
Combine MAPK10 knockdown/overexpression with antibody-based detection of multiple network components.
This approach identifies both direct and indirect effects of MAPK10 on cellular signaling networks.