Phospho-MAPK8/MAPK9/MAPK10 (T183/T183/T221) is a recombinant monoclonal antibody designed to detect phosphorylated isoforms of mitogen-activated protein kinases (MAPKs) JNK1 (MAPK8), JNK2 (MAPK9), and JNK3 (MAPK10) at residues T183 (for JNK1/JNK2) and T221 (for JNK3). These phosphorylation events occur in the activation loop of the kinase domain and are critical for enzymatic activity during cellular stress responses, apoptosis, and differentiation .
The antibody is produced by immunizing rabbits with synthetic peptides derived from human phospho-MAPK8/9/10, followed by gene cloning into mammalian expression vectors, cell culture, and affinity chromatography purification .
JNK Activation: Phosphorylation at T183/T221 is induced by stressors like UV radiation, anisomycin, or cytokines. This antibody confirmed JNK activation in NIH 3T3 cells treated with anisomycin (WB validation) .
Apoptotic Regulation: The antibody identified phosphorylated JNK1/2/3 in apoptotic pathways involving p53/TP53 and YAP1 phosphorylation .
Western Blot: Detected bands at ~48 kDa (predicted) but observed additional bands at 40–60 kDa due to isoform-specific phosphorylation or post-translational modifications .
Immunohistochemistry: Localized active JNK in Sertoli cells and heat-stressed germ cells, linking MAPK activation to apoptosis in testicular models .
Specificity Confirmation:
Lot Consistency: Affinity purification ensures batch-to-batch reproducibility .
The generation of the phospho-MAPK8/MAPK9/MAPK10 (T183/T183/T221) recombinant monoclonal antibody involves a multi-step process. First, the genes encoding the antibody are isolated from rabbits previously immunized with a synthetic peptide derived from the human phospho-MAPK8/MAPK9/MAPK10 (T183/T183/T221) protein. These antibody genes are then cloned into expression vectors and introduced into mammalian suspension cells. The cells are cultured to produce and secrete the antibody. Subsequently, the phospho-MAPK8/MAPK9/MAPK10 (T183/T183/T221) recombinant monoclonal antibody undergoes purification using affinity chromatography to isolate it from the cell culture supernatant. Finally, the antibody's functionality is rigorously assessed through a series of tests, including ELISA, Western blotting, and immunohistochemistry, to confirm its ability to specifically interact with the human phospho-MAPK8/MAPK9/MAPK10 (T183/T183/T221) protein.
The Phospho-MAPK8/MAPK9/MAPK10 (T183/T183/T221) recombinant monoclonal antibody is an immunological reagent specifically designed to recognize the phosphorylated forms of three mitogen-activated protein kinases: MAPK8 (JNK1), MAPK9 (JNK2), and MAPK10 (JNK3). This antibody specifically targets the phosphorylated threonine residues at positions 183 (MAPK8), 183 (MAPK9), and 221 (MAPK10) located within the activation loop of these kinases. The antibody recognizes these phosphorylation sites that occur after activation of the JNK signaling pathway in response to various cellular stresses and stimuli. These phosphorylation events are critical for the functional activation of these kinases, allowing them to phosphorylate downstream substrates involved in various cellular processes including proliferation, differentiation, and apoptosis .
The Phospho-MAPK8/MAPK9/MAPK10 (T183/T183/T221) recombinant monoclonal antibody has been validated for multiple experimental applications with specific recommended dilutions:
Application | Recommended Dilution |
---|---|
Western Blot (WB) | 1:500-1:5000 |
Immunohistochemistry (IHC) | 1:50-1:200 |
These recommended dilutions have been established through rigorous testing to ensure optimal signal-to-noise ratio across different experimental contexts. For Western blotting applications, the wide dilution range allows researchers to adjust based on expression levels in their specific samples. The more concentrated dilutions for immunohistochemistry reflect the need for stronger antibody concentrations when detecting phosphorylated epitopes in fixed tissue sections .
The production of the Phospho-MAPK8/MAPK9/MAPK10 (T183/T183/T221) recombinant monoclonal antibody follows a sophisticated multi-step process:
The process begins with immunizing rabbits with a synthesized peptide derived from human phospho-MAPK8/MAPK9/MAPK10 (T183/T183/T221) protein.
Genes encoding the MAPK8/MAPK9/MAPK10 (T183/T183/T221) antibody are isolated from these immunized rabbits.
These antibody genes are subsequently cloned into specialized expression vectors.
The vectors containing the antibody genes are introduced into mammalian suspension cells.
These mammalian cells are cultured to produce and secrete the antibodies.
The antibody undergoes purification through affinity chromatography to separate it from the cell culture supernatant.
The functionality of the purified antibody is comprehensively evaluated through multiple tests including ELISA, Western blot, and immunohistochemistry to confirm its specific interaction with the human phospho-MAPK8/MAPK9/MAPK10 proteins .
This recombinant approach offers advantages over traditional monoclonal antibodies, including improved batch-to-batch consistency and enhanced specificity for the target epitopes.
Researchers employ the Phospho-MAPK8/MAPK9/MAPK10 (T183/T183/T221) antibody to investigate the dynamic activation of MAPK signaling across diverse cellular contexts. In neuroblastoma SH-SY5Y cells, researchers have documented the dose-dependent activation of JNK phosphorylation following anisomycin treatment (3-1000 nM for 30 minutes), with concurrent assessment of upstream activators MKK4 and MKK7. The antibody enables detection of phosphorylation events both in basal conditions and following pharmacological interventions with JNK inhibitors such as JNK inhibitor II (20 μM) .
Similarly, studies in hepatocytes reveal that MAPK8/9 may actually suppress basal autophagy, as evidenced by increased LC3B-II accumulation following lysosomal inhibition in MAPK8/9-deficient hepatocytes compared to controls. This suppressive effect appears to operate through MAPK8/9-mediated inhibition of the PPARA (peroxisome proliferator activated receptor alpha) nuclear receptor, which normally promotes autophagy through increased expression of autophagic genes .
These contrasting findings highlight the context-dependent nature of MAPK signaling, underscoring the importance of comprehensive experimental design incorporating multiple cell types and conditions to fully characterize pathway dynamics.
The relationship between MAPK8/9/10 and autophagy regulation exhibits remarkable context-dependency across experimental systems. In murine fibroblasts and epithelial cells, MAPK8/9 are not required for autophagy induced by starvation or MTOR inhibition. Studies using both MAPK8/9-deficient immortalized MEFs and pharmacological inhibition demonstrate that autophagy proceeds normally in these contexts even without MAPK8/9 activity. When examined under rapamycin or torin 1 treatment, which inhibit TORC1 or both TORC1/TORC2 respectively, no significant differences in autophagic flux were observed between wild-type and MAPK8/9-deficient cells .
Conversely, in primary hepatocytes, MAPK8/9 appears to function as a negative regulator of autophagy. Hepatocytes from Albumin-Cre+/− Mapk8LoxP/LoxP Mapk9LoxP/LoxP mice exhibit increased LC3B-II accumulation following lysosomal inhibition and reduced levels of the autophagic substrate SQSTM1 compared to control hepatocytes. This finding was corroborated through pharmacological inhibition studies using JNK-IN-8, a potent and selective MAPK8/9 inhibitor, which similarly enhanced autophagic flux .
These divergent findings highlight that MAPK8/9's role in autophagy is not universal but rather depends on cell type, physiological context, and potentially the specific autophagic stimulus being investigated.
The Phospho-MAPK8/MAPK9/MAPK10 (T183/T183/T221) antibody recognizes all three JNK isoforms when phosphorylated at their respective activation loop residues. While this provides comprehensive coverage of JNK pathway activation, distinguishing between individual isoforms requires additional experimental strategies. Researchers can employ complementary approaches alongside this antibody to delineate isoform-specific activation:
Using genetic models: Studies can utilize cell lines or animal models with selective knockout of specific JNK isoforms (mapk8−/−, mapk9−/−, or compound knockouts) to identify which phosphorylation signals are lost when specific isoforms are absent.
Isoform-specific immunoprecipitation: By first immunoprecipitating tagged versions of individual JNK isoforms (e.g., GFP-tagged wild-type MKK7, MKK7T66A, or MKK7T83A) and then probing with the phospho-specific antibody, researchers can assess activation of specific isoforms .
Molecular weight discrimination: Since the JNK isoforms have slightly different molecular weights (MAPK8/JNK1: 48.3 kDa for the canonical isoform), high-resolution SDS-PAGE can sometimes resolve the different isoforms when probed with this antibody .
Combined with isoform-specific antibodies: Using the phospho-antibody in parallel with antibodies that recognize total levels of specific JNK isoforms allows calculation of the phosphorylation ratio for each isoform.
These approaches have been successfully employed in studies examining the differential roles of JNK isoforms in processes like anisomycin-induced stress signaling .
Detecting phosphorylated MAPKs presents several technical challenges that researchers commonly encounter:
Rapid dephosphorylation post-lysis: Phosphorylated residues on MAPKs are susceptible to rapid dephosphorylation by cellular phosphatases that remain active during sample preparation. To mitigate this issue, researchers should incorporate phosphatase inhibitors (e.g., sodium fluoride, sodium orthovanadate, β-glycerophosphate) in lysis buffers and maintain samples at 4°C throughout processing.
Low basal phosphorylation levels: Under unstimulated conditions, the basal phosphorylation of MAPK8/9/10 is often below detection threshold. This can be addressed by using more sensitive detection methods such as enhanced chemiluminescence (ECL) systems or increasing protein loading. Additionally, comparative experiments with positive controls (e.g., anisomycin-treated cells at 300 nM for 30 minutes) can help establish detection parameters .
Temporal dynamics: MAPK phosphorylation often follows rapid and transient kinetics. Time-course experiments with multiple sampling points (e.g., 5, 15, 30, 60 minutes post-stimulation) are crucial to capture peak activation windows.
Antibody cross-reactivity: While the Phospho-MAPK8/MAPK9/MAPK10 antibody is designed for specificity, validation using MAPK8/9 knockout models as negative controls is recommended to confirm signal specificity, as demonstrated in studies with mapk8−/−mapk9−/− immortalized MEFs .
Inconsistent transfer of phosphoproteins: During Western blotting, phosphoproteins may transfer inefficiently. Optimizing transfer conditions with longer transfer times or modified buffer compositions can improve detection sensitivity.
Addressing these challenges through careful experimental design and appropriate controls enables reliable detection of phosphorylated MAPKs across experimental conditions.
When studying MAPK8/9/10 phosphorylation, incorporating appropriate controls is essential for data interpretation and validation:
Positive stimulation controls: Include samples treated with known strong activators of JNK phosphorylation such as anisomycin (300 nM, 30 min), UV radiation, or inflammatory cytokines. Research has shown that anisomycin induces dose-dependent activation of JNK phosphorylation in SH-SY5Y cells across concentrations ranging from 3 to 1000 nM .
Inhibitor controls: Include samples treated with specific JNK pathway inhibitors such as JNK inhibitor II (20 μM) or JNK-IN-8. These controls help establish signal specificity and can reveal potential feedback mechanisms, as demonstrated in studies where JNK inhibition affected not only JNK phosphorylation but also upstream kinase activity .
Genetic controls: When available, include samples from cells with genetic deletion of MAPK8/9 (mapk8−/−mapk9−/−). Studies using immortalized MEFs with compound deletion of MAPK8/9 have provided valuable negative controls for antibody specificity .
Loading controls: Include detection of total (non-phosphorylated) MAPK8/9/10 to normalize phosphorylation signals and account for variations in protein loading or expression levels between samples.
Kinase activation assessment: Include detection of upstream activators (MKK4/MKK7) and downstream substrates (c-Jun) to confirm functional pathway activation. Studies have shown coordinated phosphorylation of MKK4/MKK7 alongside JNK activation following anisomycin treatment .
Time-course controls: Include multiple time points post-stimulation to account for the potentially transient nature of phosphorylation events.
Detecting subtle changes in MAPK phosphorylation requires optimized experimental conditions and enhanced sensitivity. Based on published research methodologies, researchers can implement several strategies:
Stimulus titration: Perform careful dose-response experiments with graduated concentrations of stimuli. Research has demonstrated that anisomycin concentrations between 3-1000 nM produce detectable concentration-dependent increases in JNK phosphorylation, allowing identification of threshold activation levels .
Temporal resolution: Implement dense time-course sampling, particularly during early activation phases (e.g., 5, 10, 15, 30, 60 minutes post-stimulation) to capture transient phosphorylation events that might be missed with fewer time points.
Phosphatase inhibition: Enhance lysis buffer formulations with multiple phosphatase inhibitors including sodium fluoride (50 mM), sodium orthovanadate (1 mM), β-glycerophosphate (10 mM), and proprietary phosphatase inhibitor cocktails to preserve phosphorylation status.
Signal amplification methods: Employ enhanced chemiluminescence substrates with extended sensitivity ranges or consider fluorescent secondary antibodies with digital imaging for improved quantification of subtle signals.
Enrichment techniques: Implement phosphoprotein enrichment using metal oxide affinity chromatography (MOAC) or immunoprecipitation prior to detection to concentrate phosphorylated species from dilute samples.
Quantification methods: Utilize digital image analysis with appropriate software to perform densitometry, normalizing phospho-signals to total protein levels. Express results as fold-change relative to basal conditions rather than absolute values to better visualize subtle changes .
Statistical power: Increase experimental replication (n≥3) and apply appropriate statistical tests to distinguish genuine subtle changes from experimental noise.
These optimization strategies have been successfully employed in research examining context-dependent MAPK activation profiles across different cell types and stimulation conditions.
The interpretation of conflicting data regarding MAPK8/9 roles in autophagy requires careful consideration of multiple experimental variables and contextual factors:
Cell type specificity: Research demonstrates fundamentally different roles for MAPK8/9 in autophagy regulation depending on cell type. While MAPK8/9 appears dispensable for starvation-induced autophagy in murine fibroblasts and epithelial cells, it functions as a negative regulator in primary hepatocytes. These divergent findings suggest that cellular context profoundly influences MAPK8/9 function, likely through tissue-specific expression of regulatory partners and downstream effectors .
Experimental approach considerations: Studies showing contradictory results may employ different methodological approaches. Genetic models (MAPK8/9 knockout) may yield different results than pharmacological inhibition (JNK inhibitors) due to compensatory mechanisms or off-target effects. For example, research using JNK-IN-8 in wild-type hepatocytes showed increased autophagic flux, corroborating findings from genetic MAPK8/9 deletion models .
Autophagy stimulus specificity: MAPK8/9 involvement may depend on the autophagy trigger being studied. Research shows that while MAPK8/9 is dispensable for starvation or MTOR inhibition-induced autophagy, its role in other contexts (e.g., stress-induced autophagy) may differ substantially. Experiments with rapamycin and torin 1 demonstrate MAPK8/9-independent autophagy induction, whereas other contexts might reveal MAPK8/9 dependency .
Mechanistic integration: Researchers should consider that MAPK8/9 may regulate autophagy through diverse mechanisms depending on context. In hepatocytes, MAPK8/9 suppresses autophagy through repression of PPARA-mediated transcription of autophagic genes, resembling the mechanism observed in neurons where MAPK8/9/10 represses autophagic gene expression .
Temporal considerations: Conflicting results may reflect differences in the timing of observations, as MAPK signaling and autophagy are both dynamic processes with complex temporal regulation.
These considerations highlight that MAPK8/9 involvement in autophagy is context-dependent and more complex than previously recognized. Rather than viewing conflicting data as contradictory, researchers should interpret these findings as revealing the multifaceted nature of MAPK signaling in autophagy regulation across different physiological and cellular contexts.
Multiple factors influence the specificity and sensitivity of phospho-MAPK8/9/10 detection in experimental settings:
Antibody characteristics: The Phospho-MAPK8/MAPK9/MAPK10 (T183/T183/T221) recombinant monoclonal antibody demonstrates high specificity for activation loop phosphorylation of all three JNK isoforms. The recombinant nature of this antibody contributes to batch-to-batch consistency, reducing experimental variability compared to conventional antibodies .
Phosphorylation kinetics: MAPK phosphorylation events are often transient, making detection timing critical. Research has shown that anisomycin treatment (300 nM for 30 minutes) produces robust phosphorylation signals, while signals at earlier or later time points may be significantly weaker .
Cross-reactivity with related MAPKs: While the antibody specifically targets phosphorylated MAPK8/9/10, potential cross-reactivity with phosphorylated p38 or ERK MAPKs should be considered, especially in contexts where multiple MAPK pathways are simultaneously activated. Studies examining anisomycin-induced signaling have evaluated activation loop phosphorylation of JNK alongside p38 and ERK to distinguish pathway-specific responses .
Background phosphorylation levels: Basal phosphorylation of MAPKs varies considerably between cell types and culture conditions. Primary cells often exhibit different basal phosphorylation profiles compared to immortalized cell lines, requiring optimization of detection parameters for each experimental system .
Signal amplification methods: The choice of detection system (chemiluminescence, fluorescence, colorimetric) significantly impacts sensitivity. Enhanced chemiluminescence systems can improve detection of low-abundance phosphorylation events but may saturate with strong signals, compromising quantification accuracy.
Sample preparation variables: Phosphorylation status can be significantly affected by sample handling, including lysis method, buffer composition, temperature, and time between cell harvesting and analysis. Research protocols typically emphasize rapid processing at cold temperatures with phosphatase inhibitors to preserve phosphorylation status .
Awareness of these factors allows researchers to optimize experimental conditions for reliable and reproducible phospho-MAPK8/9/10 detection across different experimental contexts.
The pronounced context-dependency of MAPK signaling necessitates thoughtful experimental design considerations:
Recent advances in MAPK8/9/10 research methodologies reflect a shift toward more nuanced, systems-level approaches to understanding these complex signaling networks. The development of recombinant monoclonal antibodies like the Phospho-MAPK8/MAPK9/MAPK10 (T183/T183/T221) antibody represents a significant improvement in detection specificity and reproducibility compared to earlier polyclonal reagents . This technical advancement parallels conceptual shifts in how researchers approach MAPK signaling.
Emerging trends include integrated multi-omics approaches that combine phosphoproteomics with transcriptomics and metabolomics to provide comprehensive pathway activation profiles. Additionally, single-cell analysis technologies are increasingly being applied to understand cell-to-cell variability in MAPK activation, revealing heterogeneity that was previously masked in population-level studies. The growing implementation of CRISPR/Cas9-mediated genome editing has facilitated more precise genetic models, allowing researchers to investigate isoform-specific functions with unprecedented resolution.
Perhaps most significantly, the field is moving away from linear pathway models toward network-based conceptualizations that account for the context-dependency observed in MAPK signaling. This is exemplified by studies demonstrating that MAPK8/9 functions as an autophagy inhibitor in hepatocytes through PPARA suppression, while being dispensable for autophagy in other cell types . These methodological and conceptual advances promise to resolve longstanding contradictions in the literature and provide more physiologically relevant insights into MAPK8/9/10 biology.
Future research directions that could significantly advance our understanding of MAPK8/9/10 signaling include:
Tissue-specific signaling networks: Given the demonstrated context-dependency of MAPK8/9/10 function, comprehensive mapping of tissue-specific MAPK signaling networks would provide valuable insights. This could involve comparative phosphoproteomic analyses across diverse primary cell types under standardized conditions to identify tissue-specific substrates and regulatory mechanisms .
Subcellular compartment-specific signaling: Investigation of how MAPK8/9/10 signaling differs across subcellular compartments (nucleus, cytoplasm, mitochondria, endoplasmic reticulum) could reveal localized functions that contribute to the complexity of MAPK biology. Research has already established differential activities of MAPK8/9 in cytoplasmic versus nuclear compartments .
Integration with metabolic regulation: The emerging connection between MAPK8/9 and metabolic regulators like PPARA suggests an underexplored role in metabolic homeostasis. Future research investigating how MAPK8/9/10 integrates with metabolic signaling networks could reveal novel therapeutic targets for metabolic disorders .
Temporal signaling dynamics: Development of real-time MAPK activity biosensors would enable more precise characterization of signaling dynamics at single-cell resolution, potentially revealing oscillatory patterns or threshold effects that are obscured in conventional endpoint analyses.
Therapeutic targeting strategies: Given the complex and context-dependent nature of MAPK8/9/10 signaling, future therapeutic approaches might benefit from tissue-specific or activation-state-specific targeting strategies rather than global inhibition. Research developing conformation-specific inhibitors that target activated versus inactive MAPK forms could provide more nuanced therapeutic options.