MAPK8 antibodies are immunological reagents designed to detect mitogen-activated protein kinase 8 (MAPK8/JNK1), a 48.3 kDa protein encoded by the MAPK8 gene in humans . These antibodies target specific epitopes, such as the phosphorylated tyrosine residue Y185 , and are critical for elucidating MAPK8's involvement in signaling pathways linked to stress responses, autophagy, and apoptosis .
MAPK8 antibodies have revealed conflicting roles in autophagy. While some studies suggest MAPK8 promotes starvation-induced autophagy via BCL2 phosphorylation , others show MAPK8/9 inhibition increases autophagic flux in hepatocytes .
In mapk8⁻/⁻ mapk9⁻/⁻ MEFs, MAPK8/9 activation alone did not induce LC3B-II conversion, indicating context-dependent roles .
MAPK8 antibodies help identify its involvement in TNF-α-induced apoptosis and UV radiation-induced cell death .
Interactions with PPARA and transcriptional repression of autophagy-related genes highlight its role in metabolic regulation .
Validation Methods: Antibodies are tested in WB, IHC, and ELISA using positive/negative controls to ensure specificity .
Example Validation: Anti-JNK1/2/3 (Y185) MAPK8 Antibody (Boster Bio #A02608Y185) showed robust reactivity in human breast carcinoma tissues at 1:100 dilution .
Storage: Stable at -20°C long-term or 4°C for short-term use .
MAPK8 (also known as JNK1) is a serine/threonine-protein kinase implicated in diverse cellular processes, including proliferation, differentiation, migration, transformation, 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 MAPK8/JNK1. Subsequently, MAPK8/JNK1 phosphorylates numerous transcription factors, primarily AP-1 components (JUN, JDP2, and ATF2), thereby modulating AP-1 transcriptional activity. Additional functions of MAPK8 include:
JNK1 isoforms exhibit distinct binding patterns: β-1 preferentially binds c-Jun, while α-1, α-2, and β-2 demonstrate similar low-level binding to both c-Jun and ATF2. However, binding does not correlate with phosphorylation efficiency, which is comparable across all isoforms.
The following studies highlight the diverse roles of JNK1 in various biological processes:
MAPK8 (mitogen-activated protein kinase 8) is a serine/threonine protein kinase also known as JNK1, SAPK1, JNK-46, JNK1A2, SAPK1c, JNK21B1/2, and PRKM8. It belongs to the MAPK superfamily of stress-activated protein kinases and is expressed as multiple isoforms due to differential mRNA splicing, with JNK1 and JNK2 being the predominant forms . MAPK8 is expressed in various tissue types and has a molecular weight of approximately 48.3 kDa, with the canonical human protein consisting of 427 amino acid residues .
MAPK8 functions as a crucial mediator in multiple cellular processes including:
Signal transduction from cell surface to nucleus
Cellular proliferation, differentiation, and migration
Transcription regulation and development
Apoptosis and cell death pathways
Response to stress and inflammatory signals
Insulin resistance and obesity mediation
Activation in response to misfolded proteins in the endoplasmic reticulum
When activated, MAPK8 phosphorylates various substrates including transcription factors like c-Jun, altering their activities and subcellular localization.
When selecting a MAPK8 antibody, researchers should consider:
Target specificity: Determine whether you need an antibody that specifically recognizes MAPK8/JNK1 or one that recognizes multiple JNK family members (JNK1/2/3)
Phosphorylation state: Select antibodies that detect total MAPK8 or phosphorylated forms (pT183/pY185)
Host species compatibility: Ensure minimal cross-reactivity with samples from other species
Application suitability: Verify antibody validation for your specific application (WB, IHC, IF, ELISA, IP)
Clone type: Consider whether monoclonal (higher specificity) or polyclonal (broader epitope recognition) is more appropriate
For optimal results, review published literature using your antibody of interest to confirm its reliability in similar experimental contexts.
Thorough validation of MAPK8 antibodies should include:
Western blotting with positive controls: Use cell lysates known to express MAPK8 (e.g., K562 cells) to confirm detection at the expected molecular weight (approximately 44/52 kDa)
Knockout/knockdown controls: Compare antibody reactivity in wild-type vs. MAPK8-deficient samples
Phospho-specific validation: For phospho-specific antibodies, compare untreated vs. stimulated samples and/or phosphatase-treated controls
Cross-reactivity testing: Test against closely related proteins (JNK2/JNK3) to ensure specificity
Peptide competition assays: Pre-incubate antibody with immunizing peptide to demonstrate signal reduction
Documenting these validation steps is essential for high-quality research publications and reproducibility.
For optimal Western blot results with MAPK8 antibodies:
Sample preparation:
Use fresh cell/tissue lysates in RIPA or NP-40 buffer with protease/phosphatase inhibitors
For phospho-JNK detection, stimulate cells with appropriate stressors (UV, cytokines, etc.)
Protein separation:
Use 10-12% SDS-PAGE gels for optimal resolution
Load 20-40 μg of total protein per lane
Include molecular weight markers spanning 40-60 kDa range
Transfer and detection:
Transfer to PVDF membranes (preferred over nitrocellulose for phospho-epitopes)
Block with 5% BSA in TBST (not milk for phospho-antibodies)
Primary antibody dilution: typically 1:1000-1:2000 in blocking buffer
Incubate overnight at 4°C with gentle agitation
Secondary antibody: 1:5000-1:10000, 1 hour at room temperature
Expected results:
For optimal IHC/IF with MAPK8 antibodies:
Sample preparation:
Fixation: 4% paraformaldehyde (10% formalin) for tissues
For IF on cultured cells: 4% PFA for 15 minutes at room temperature
Antigen retrieval (for fixed tissues):
Heat-induced epitope retrieval in citrate buffer (pH 6.0)
For phospho-epitopes: use EDTA buffer (pH 8.0)
Blocking and antibody incubation:
Block with 5-10% normal serum from secondary antibody host species
Primary antibody dilution: typically 1:100-1:500
Incubation time: 1-2 hours at room temperature or overnight at 4°C
Detection systems:
For IHC: HRP-conjugated secondary + DAB substrate
For IF: fluorophore-conjugated secondary antibody (avoid overlapping wavelengths if co-staining)
Controls:
Recent research identified MAPK8 as a key biomarker in intervertebral disc degeneration (IDD). A 2023 study by Frontiers in Immunology showed:
MAPK8's role in IDD:
Recommended methodology for studying MAPK8 in IDD:
Bioinformatic approaches: Start with DEG analysis followed by MCODE plugin for hub gene identification
Animal models: Use needle puncture technique to induce IDD in rats
Validation techniques: RT-qPCR for mRNA expression levels, comparing with control markers (aggrecan and COL-2)
Imaging validation: X-ray, MRI, and H&E staining of NP tissue sections
Mechanistic studies: Examine MAPK8's relationship with autophagy pathways and immune cell infiltration
The study demonstrated that unlike other identified hub genes, MAPK8 expression was consistently elevated in IDD, suggesting its potential as a therapeutic target for this condition.
A 2024 Nature study revealed MAPK8's important role in lung adenocarcinoma (LUAD) progression:
These findings suggest that targeting the miR-147b/DUSP8/MAPK8 axis could provide novel approaches for lung cancer treatment.
Research on MAPK8's role in autophagy presents some contradictory findings:
Supporting evidence for MAPK8's essential role:
Initial studies reported that MAPK8/JNK1 (but not MAPK9/JNK2) is required for starvation-induced autophagy
Proposed mechanism: BCL2 phosphorylation (on Thr69, Ser70, and Ser87) by MAPK8 disrupts BCL2-BECN1 interaction
This initiates BECN1-dependent autophagy
Contradicting evidence:
Some studies found MAPK8/JNK1 and MAPK9/JNK2 are not required for autophagy caused by starvation or MTOR inhibition in murine fibroblasts and epithelial cells
MAPK9/JNK2 may also play contributing roles in autophagy
In primary hepatocytes, MAPK8/9 appears to suppress rather than promote autophagic flux
Pharmacological inhibition with JNK-IN-8 increased autophagic flux in some contexts
Reconciling the contradictions:
Researchers investigating MAPK8's role in autophagy should carefully design experiments with appropriate controls and consider multiple cell types to address these contradictions.
When developing multiplexed immunoassays for MAPK8:
Antibody pair selection:
Ensure epitopes recognized by capture and detection antibodies do not overlap
Validate antibody pairs to confirm they don't interfere with each other's binding
Consider using monoclonal antibodies with defined epitopes for highest specificity
Cross-reactivity mitigation:
Test for cross-reactivity between antibodies in the multiplex panel
Implement extensive blocking procedures to minimize non-specific binding
Use species-matched negative controls for each antibody
Signal separation strategies:
For fluorescence-based detection: Choose fluorophores with minimal spectral overlap
For chromogenic detection: Ensure signal separation through distinct substrate reactions
Consider sequential detection protocols if cross-reactivity cannot be eliminated
Validation approaches:
Data normalization:
Include internal reference proteins for normalization
Account for potential signal crosstalk during data analysis
Consider using machine learning approaches for signal deconvolution in complex samples
Non-specific binding is a common challenge with MAPK8 antibodies. Here are potential causes and solutions:
| Problem | Potential Causes | Solutions |
|---|---|---|
| Multiple bands in Western blot | - Detection of multiple MAPK8 isoforms - Cross-reactivity with JNK2/JNK3 - Sample degradation | - Confirm band pattern with positive control - Use isoform-specific antibodies - Include protease inhibitors in lysis buffer |
| High background staining | - Insufficient blocking - Excessive antibody concentration - Non-specific binding to Fc receptors | - Optimize blocking conditions (5% BSA or 5% milk) - Titrate antibody concentrations - Add species-matched IgG to blocking buffer |
| False positive signals | - Endogenous peroxidase activity (IHC) - Autofluorescence (IF) - Endogenous biotin (if using avidin-biotin systems) | - Include hydrogen peroxide quenching step - Use autofluorescence quenchers - Use biotin blocking kit for avidin-biotin detection |
| Inconsistent results | - Batch-to-batch variability - Antibody degradation - Inconsistent sample preparation | - Note lot numbers and validate each new lot - Aliquot antibodies to minimize freeze-thaw cycles - Standardize sample collection and processing |
For phospho-specific MAPK8 antibodies, additional considerations include maintaining phosphatase inhibitors throughout sample preparation and optimizing stimulation conditions to ensure robust phosphorylation .
Distinguishing between JNK family members requires careful antibody selection and validation:
Antibody selection strategies:
Use antibodies raised against unique peptide sequences specific to each JNK isoform
Select antibodies that have been specifically validated against all three JNK proteins
Consider using antibodies designed to recognize specific splice variants
Validation approaches:
Test antibodies on samples with known expression patterns of specific JNK isoforms
Use genetic models (knockouts or knockdowns) for each JNK family member
Employ overexpression systems with tagged versions of each JNK for positive controls
Technical considerations:
Western blot can distinguish JNK isoforms by molecular weight differences:
JNK1/MAPK8: 46 kDa (p46) and 54 kDa (p54)
JNK2/MAPK9: 55 kDa
JNK3/MAPK10: 46 kDa
For IHC/IF, validate specificity through co-staining with isoform-specific markers
Consider using RNA methods (RT-PCR, RNA-seq) as complementary approaches to confirm protein findings
Common pitfalls:
Many commercial antibodies show cross-reactivity between JNK family members
Post-translational modifications can alter mobility in gels
Tissue-specific expression patterns may complicate interpretation (JNK3 is predominantly expressed in brain)
Single-cell analysis of MAPK8 in heterogeneous tissues presents both challenges and opportunities:
Single-cell immunostaining approaches:
Multiplex immunofluorescence with MAPK8 antibodies and cell-type markers
Mass cytometry (CyTOF) using metal-conjugated MAPK8 antibodies
Imaging mass cytometry for spatial context within tissues
Proximity ligation assays to detect MAPK8 interactions at single-cell level
Single-cell sequencing integration:
Correlate protein measurements with transcriptomic data using CITE-seq
Apply computational methods to integrate antibody-based measurements with RNA profiles
Use pseudotime analysis to track MAPK8 activity changes during cellular processes
Spatial considerations:
Implement multiplexed immunohistochemistry with MAPK8 antibodies
Apply digital spatial profiling technologies
Correlate MAPK8 status with tissue microenvironment features
Challenges and solutions:
Recent applications in intervertebral disc degeneration research have shown that MAPK8 expression correlates with specific immune cell infiltration patterns, demonstrating the value of examining this signaling molecule in a cell type-specific context .
MAPK8 antibodies are increasingly important in therapeutic development:
Target validation:
Use MAPK8 antibodies to confirm target engagement in drug development
Apply for pharmacodynamic biomarker development
Validate functional consequences of MAPK8 pathway modulation
Patient stratification:
Develop companion diagnostics using MAPK8 antibodies
Identify patient subgroups likely to respond to JNK pathway modulators
Monitor MAPK8 activation status as a predictive biomarker
Therapeutic monitoring:
Track MAPK8 phosphorylation status during treatment
Develop multiplexed assays to monitor multiple nodes in the pathway
Use in clinical trials to establish pharmacokinetic/pharmacodynamic relationships
Emerging therapeutic areas:
Technological advances:
Antibody-drug conjugates targeting MAPK8-expressing cells
Intrabodies directed against active conformations of MAPK8
Nanobody development for improved tissue penetration and target access
These emerging applications highlight the growing importance of high-quality, well-validated MAPK8 antibodies in both basic research and translational medicine contexts.
To ensure reliable results with MAPK8 antibodies, implement these quality control measures:
Documentation and record-keeping:
Maintain detailed records of antibody source, catalog number, lot number, and validation data
Document all experimental conditions, including incubation times, temperatures, and buffer compositions
Create a laboratory antibody validation database
Routine validation:
Test each new antibody lot against a reference standard
Include appropriate positive and negative controls in every experiment
Periodically reassess antibody performance, especially after prolonged storage
Application-specific controls:
Western blot: Include molecular weight markers and positive control lysates
IHC/IF: Include known positive tissues and secondary-only controls
IP: Perform IgG control pulldowns alongside target pulldowns
Storage and handling:
Aliquot antibodies to minimize freeze-thaw cycles
Store according to manufacturer recommendations (typically -20°C)
Monitor for signs of degradation or contamination
Reporting standards:
Implementing these quality control measures will significantly enhance reproducibility and reliability of MAPK8 antibody-based experiments.
When designing experiments to study MAPK8 across different cellular contexts:
Cell type considerations:
Account for tissue-specific expression patterns of MAPK8 isoforms
Consider basal activation state of JNK pathway in different cell types
Adapt lysis conditions to cell type (adherent vs. suspension, primary vs. cell line)
Stimulus selection:
Choose physiologically relevant stimuli for the cell type under study
Establish appropriate time courses (JNK activation can be transient)
Consider combinatorial stimuli to mimic complex in vivo conditions
Detection strategy optimization:
Select antibodies validated in your specific cell type
Optimize fixation conditions for each cell type (especially for primary cells)
Consider subcellular localization analysis (nuclear translocation upon activation)
Context-dependent interpretation:
Translation between models: