CUSABIO designed the vector clones for the expression of a recombinant MAP2K1 antibody in mammalian cells. The vector clones were obtained by inserting the MAP2K1 antibody heavy and light chains into the plasma vectors. The recombinant MAP2K1 antibody was purified from the culture medium through affinity-chromatography. It can be used to detect MAP2K1 protein from Human in the ELISA, WB.
The phospho-MAP2K1 (T292) antibody can detect the MAP2K1 only when phosphorylated at T292. MAP2K1, also called MEK1, functions immediately upstream of MAPK in the MAPK signaling pathway. Phosphorylation of T292 on MAP2K1 by activated ERK is required for the formation of ternary complex PTEN/MAGI1/MAP2K1. MAP2K1 mutations have been found in several human malignancies, particularly melanoma, hairy cell leukemia, and lung adenocarcinoma.
CUSABIO has engineered vector clones for the expression of a recombinant MAP2K1 antibody in mammalian cells. These clones were generated by inserting the MAP2K1 antibody heavy and light chains into suitable plasma vectors. The recombinant MAP2K1 antibody was subsequently purified from the culture medium using affinity-chromatography. This antibody is designed for the detection of MAP2K1 protein from Human in ELISA and Western blotting applications.
The phospho-MAP2K1 (T292) antibody exhibits specificity for MAP2K1 only when phosphorylated at T292. MAP2K1, also known as MEK1, plays a pivotal role as an upstream regulator of MAPK within the MAPK signaling pathway. Phosphorylation of T292 on MAP2K1 by activated ERK is essential for the formation of a ternary complex composed of PTEN, MAGI1, and MAP2K1. Notably, mutations in MAP2K1 have been implicated in various human malignancies, particularly melanoma, hairy cell leukemia, and lung adenocarcinoma.
MAP2K1 (also known as MEK1) is a dual specificity protein kinase that serves as a critical component of the MAP kinase signal transduction pathway. The activation of this pathway is initiated by the binding of extracellular ligands, such as growth factors, cytokines, and hormones, to their corresponding cell-surface receptors. This binding event triggers the activation of RAS, which in turn activates RAF1. RAF1 then further activates the dual-specificity protein kinases MAP2K1/MEK1 and MAP2K2/MEK2. Both MAP2K1/MEK1 and MAP2K2/MEK2 are specifically involved in the MAPK/ERK cascade. They catalyze the phosphorylation of a threonine and a tyrosine residue within a Thr-Glu-Tyr sequence located in the extracellular signal-regulated kinases MAPK3/ERK1 and MAPK1/ERK2. This phosphorylation event leads to the activation of these kinases, allowing for further signal transduction within the MAPK/ERK cascade.
MAP2K1/MEK1 also activates BRAF in a KSR1 or KSR2-dependent manner. Binding to KSR1 or KSR2 releases the inhibitory intramolecular interaction between the KSR1 or KSR2 protein kinase and N-terminal domains, which promotes KSR1 or KSR2-BRAF dimerization and subsequent activation of BRAF. Depending on the cellular context, the MAPK/ERK cascade orchestrates a wide range of biological functions, including cell growth, adhesion, survival, and differentiation. These functions are primarily mediated through the regulation of transcription, metabolism, and cytoskeletal rearrangements. One target of the MAPK/ERK cascade is peroxisome proliferator-activated receptor gamma (PPARG), a nuclear receptor that promotes differentiation and apoptosis. MAP2K1/MEK1 has been shown to export PPARG from the nucleus. The MAPK/ERK cascade also participates in the regulation of endosomal dynamics, including lysosome processing and endosome cycling through the perinuclear recycling compartment (PNRC), as well as the fragmentation of the Golgi apparatus during mitosis.
T292 phosphorylation on MAP2K1 (also known as MEK1) represents a critical regulatory mechanism within the MAPK/ERK signaling cascade. This phosphorylation event occurs as part of a negative feedback loop where activated ERK phosphorylates MEK1 at T292, which subsequently interferes with PAK-mediated phosphorylation of MEK1 at S298 . This negative feedback mechanism helps maintain homeostatic control of MAPK pathway activation. The T292 phosphorylation acts as a molecular switch that modulates signal duration and intensity by attenuating continued pathway activation. Understanding this phosphorylation event is crucial for interpreting experimental results when studying MAPK pathway dynamics, especially in cancer research where this pathway is frequently dysregulated .
The Phospho-MAP2K1 (T292) antibody specifically recognizes MEK1 phosphorylated at threonine 292, distinguishing it from antibodies targeting other phosphorylation sites such as S218/S222 (activation loop phosphorylation) or T386 (another ERK-mediated feedback phosphorylation site) . This specificity allows researchers to selectively monitor the negative feedback regulation of MEK1 by ERK, rather than its activation state. The antibody has been rigorously validated using both wild-type MEK1 and T292A mutant proteins, confirming its specificity for the phosphorylated T292 residue . Unlike antibodies targeting the activation loop phosphorylation (which indicate MEK1 activation), the T292 phospho-antibody provides insight into pathway regulation and signal termination mechanisms, offering complementary information when used alongside other phospho-specific antibodies in signaling studies .
The Phospho-MAP2K1 (T292) Recombinant Antibody has been validated for specific research applications through rigorous testing. Current validated applications include:
Validation has been performed using recombinant wild-type and mutant (T292A) MAP2K1 proteins, with and without co-expression of MAP kinase, demonstrating specific immunolabeling of the phosphorylated form . The antibody has shown reactivity with human samples and potentially cross-reacts with bovine, dog, mouse, primate, and rat samples due to sequence conservation around the T292 site . It's important to note that optimal dilutions should be determined by each laboratory for specific applications and experimental conditions .
Designing appropriate controls is critical for accurately interpreting results with Phospho-MAP2K1 (T292) antibody. A comprehensive control strategy should include:
Positive Control: Lysates from cells treated with agents known to induce ERK activation (e.g., EGF, PMA) should show increased T292 phosphorylation due to the negative feedback mechanism .
Negative Controls:
Loading Controls: Total MEK1 antibody should be used on parallel blots or after stripping to normalize phospho-signal to total protein levels .
Pathway Controls: Monitoring ERK activation (phospho-ERK1/2) in parallel is essential since T292 phosphorylation is ERK-dependent .
A technically robust experiment demonstrated in validation studies includes expressing wild-type and T292A mutant MEK1 with and without MAP kinase co-expression, clearly showing that the antibody only detects the phosphorylated form when both wild-type MEK1 and active MAP kinase are present . This approach definitively confirms antibody specificity and is recommended for researchers validating the antibody in their experimental systems.
Optimal detection of MAP2K1 T292 phosphorylation requires careful consideration of stimulation conditions, as this phosphorylation represents a negative feedback mechanism following ERK activation. Based on the MAPK pathway biology:
Temporal Considerations: T292 phosphorylation typically occurs after initial MEK activation and subsequent ERK activation. Time-course experiments are recommended:
Effective Stimulants:
Cell Types: Cell lines with robust MAPK pathway activity (e.g., HEK293, NIH3T3, various cancer cell lines) typically show stronger T292 phosphorylation signals .
Inhibitor Studies: Using MEK inhibitors (U0126, PD0325901) or ERK inhibitors (SCH772984) can help establish the dependency of T292 phosphorylation on pathway activation. These should eliminate the phospho-signal, confirming specificity .
The detection is optimized when cells are lysed in buffers containing phosphatase inhibitors (sodium fluoride, sodium orthovanadate, and phosphatase inhibitor cocktails) to preserve the phosphorylation status during sample preparation .
Preserving MAP2K1 T292 phosphorylation during protein extraction is critical for accurate detection and quantification. Recommended extraction protocols include:
Lysis Buffer Composition:
Base buffer: 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% NP-40 or Triton X-100
Critical phosphatase inhibitors:
10 mM sodium fluoride
2 mM sodium orthovanadate (freshly activated)
10 mM β-glycerophosphate
Commercial phosphatase inhibitor cocktail (1X)
Protease inhibitors: PMSF (1 mM) and protease inhibitor cocktail (1X)
Extraction Procedure:
Sample Handling:
Tissue Samples:
The effectiveness of phosphorylation preservation can be assessed by comparing phospho-ERK levels in the same samples, as both modifications are phosphatase-sensitive and occur in the same signaling pathway .
Phospho-MAP2K1 (T292) antibodies provide a valuable tool for investigating the complex cross-talk between MAPK and other signaling pathways. This phosphorylation site serves as a critical node where multiple pathways converge to regulate MAPK signaling:
MAPK-PAK Pathway Cross-talk: T292 phosphorylation by ERK interferes with PAK-mediated phosphorylation of MEK1 at S298, creating a phospho-switch mechanism. Researchers can use the T292 phospho-antibody alongside a S298 phospho-antibody to monitor this regulatory cross-talk in real-time following various stimuli .
Rac/Cdc42 Signaling Integration: PAK is activated downstream of Rac/Cdc42 small GTPases, so T292 phosphorylation represents a point where Rac/Cdc42 signaling intersects with the MAPK pathway. Experiments combining Rac/Cdc42 activators with MAPK pathway stimulants can reveal how these pathways counter-regulate each other through MEK1 phosphorylation, quantifiable via immunoblotting with the T292 antibody .
PI3K-AKT-MAPK Cross-regulation: Multiple studies suggest interactions between PI3K/AKT and MAPK pathways. Researchers can design experiments using specific inhibitors of PI3K (LY294002), AKT (MK2206), and MAPK pathway components while monitoring T292 phosphorylation to map pathway dependencies .
Experimental Approach for Cross-talk Studies:
Stimulate cells with pathway-specific activators individually and in combination
Quantify T292 phosphorylation alongside other pathway-specific phosphorylation events
Apply specific pathway inhibitors to define the hierarchy of regulation
Monitor temporal dynamics of phosphorylation events to establish causality
This approach has revealed important insights in cancer research, where alterations in feedback mechanisms contribute to therapy resistance and pathway rewiring, particularly in melanoma where MAP2K1/2 mutations occur in approximately 8% of patients .
MAP2K1 T292 phosphorylation has significant implications in cancer research, particularly regarding therapeutic resistance mechanisms:
Researchers can use the Phospho-MAP2K1 (T292) antibody to assess these mechanisms in patient samples and preclinical models, potentially guiding personalized treatment strategies based on pathway feedback status .
The phosphorylation status of MAP2K1 at T292 provides critical insights into how cells respond to targeted MAPK pathway inhibitors:
Predictive Biomarker for Drug Response: T292 phosphorylation status prior to treatment may indicate the degree of feedback regulation already present in the tumor cells, potentially predicting initial response to MAPK pathway inhibitors. Cells with abnormally low T292 phosphorylation despite high pathway activity may indicate disrupted feedback mechanisms and potentially poorer response to single-agent therapy .
Dynamic Biomarker During Treatment: Monitoring T292 phosphorylation during treatment reveals important adaptive responses:
Initial decrease in T292 phosphorylation after MEK or RAF inhibitor treatment indicates successful pathway inhibition
Rapid recovery of T292 phosphorylation despite continued treatment suggests pathway reactivation and developing resistance
Sustained suppression of T292 phosphorylation correlates with durable response
Mechanistic Insights from Combination Studies: Research using phospho-specific antibodies has revealed that:
MEK inhibitors block downstream ERK activation, reducing T292 phosphorylation
RAF inhibitors in BRAF-mutant cells initially decrease T292 phosphorylation
In RAS-mutant cells, RAF inhibitors can paradoxically increase T292 phosphorylation due to RAF dimerization
Combining RAF and MEK inhibitors more effectively suppresses T292 phosphorylation and improves response durability
Correlation with Clinical Outcomes: While more clinical validation is needed, preclinical data suggests that persistent T292 phosphorylation despite MAPK pathway inhibitor treatment correlates with poorer response. Analysis of patient-derived samples before and after treatment progression using Phospho-MAP2K1 (T292) antibodies can provide valuable biomarker data for predicting treatment efficacy .
Researchers can implement regular monitoring of T292 phosphorylation alongside other pathway markers in drug response studies to gain deeper mechanistic understanding of treatment effects and resistance development .
Researchers commonly encounter several technical challenges when working with Phospho-MAP2K1 (T292) antibodies. Here are evidence-based solutions to these issues:
Low Signal Intensity:
Cause: Rapid dephosphorylation during sample preparation or low baseline phosphorylation
Solution: Enhance phosphatase inhibitor cocktail (double sodium orthovanadate to 4mM); pre-treat cells with phosphatase inhibitors (calyculin A, okadaic acid) for 15-30 minutes before lysis; optimize stimulation conditions to maximize T292 phosphorylation
High Background or Non-specific Bands:
Cause: Antibody concentration too high; insufficient blocking; cross-reactivity
Solution: Optimize antibody dilution (try 1:2000-1:5000); increase blocking time/concentration (5% BSA often works better than milk for phospho-epitopes); add 0.1% Tween-20 to antibody diluent; confirm specificity using T292A mutant as negative control
Poor Reproducibility Between Experiments:
Cause: Variable phosphorylation status; inconsistent sample preparation
Solution: Standardize cell density and stimulation protocol; prepare fresh lysis buffers for each experiment; minimize time between lysis and denaturation; consider using phospho-protein stabilizing buffers commercially available
Weak Signal in Tissue Samples:
Antibody Performance Validation:
The validation methodology shown in multiple sources demonstrates that appropriate controls can distinguish specific signal from artifacts, with clear detection of the ~45 kDa phosphorylated MAP2K1 protein only in samples with active MAP kinase and intact T292 phosphorylation site .
Optimizing multiplexed detection of different MAP2K1 phosphorylation sites requires careful experimental design to obtain comprehensive pathway analysis. Here's a systematic approach:
Antibody Selection and Validation:
Choose antibodies raised in different host species (e.g., rabbit anti-phospho-T292, mouse anti-phospho-S218/S222) to enable simultaneous detection
Validate each antibody individually using appropriate controls (phosphatase treatment, site-specific mutants)
Test for cross-reactivity between antibodies to ensure specific detection
Sequential Immunoblotting Approach:
Multi-color Fluorescent Western Blotting:
Sample Preparation Optimization:
Different phosphorylation sites may have different sensitivities to phosphatases
Use a comprehensive phosphatase inhibitor mixture:
Inhibitor | Concentration | Target Phosphatases |
---|---|---|
Sodium fluoride | 50 mM | Serine/threonine phosphatases |
Sodium orthovanadate | 2-5 mM | Tyrosine phosphatases |
β-glycerophosphate | 10 mM | Serine/threonine phosphatases |
Sodium pyrophosphate | 5 mM | Serine/threonine phosphatases |
EDTA/EGTA | 5 mM | Metallo-phosphatases |
Commercial cocktail | 1X | Broad spectrum |
Alternative Technologies for Multiplexed Detection:
Researchers should note that different phosphorylation sites may have different temporal dynamics after stimulation. T292 phosphorylation (feedback mechanism) typically occurs after activation loop (S218/S222) phosphorylation, so time-course experiments are essential for comprehensive pathway analysis .
Proper quantification and statistical analysis of phospho-specific antibody data is crucial for deriving meaningful biological interpretations. Here's a comprehensive approach:
Quantification Methods:
Densitometry Analysis:
Normalization Hierarchy (from most to least accurate):
Phospho-protein/Total target protein ratio from same membrane (multiplex fluorescent detection)
Phospho-protein/Total target protein ratio from stripped and reprobed membrane
Phospho-protein/Total target protein ratio from parallel membranes
Phospho-protein/Loading control ratio (least reliable for phosphorylation studies)
Statistical Analysis Framework:
For Time-Course Studies:
For Dose-Response Studies:
For Multiple Treatment Comparisons:
Biological Replication Requirements:
Validation Through Orthogonal Methods:
Data Presentation Best Practices:
Meaningful interpretation requires correlation with functional outcomes, so researchers should connect phosphorylation data with downstream biological effects and consider pathway modeling approaches for systems-level understanding .
MAP2K1 T292 phosphorylation status represents a potentially valuable biomarker for patient stratification in both targeted therapy and immunotherapy contexts. Current evidence suggests several promising applications:
Predictive Biomarker Development:
Research has shown that MAP2K1/2 mutations may correlate with response to immune checkpoint inhibitors, particularly CTLA-4 blockade therapy in melanoma patients .
T292 phosphorylation status could serve as a functional readout of these mutations' effects on pathway regulation.
Immunohistochemistry protocols using phospho-T292 antibodies on patient biopsies could be developed for clinical implementation .
Integration with Genetic Biomarkers:
Combining T292 phosphorylation analysis with genetic testing for MAP2K1/2 mutations provides complementary information:
Biomarker Combination | Potential Clinical Interpretation |
---|---|
MAP2K1/2 mutation (+) / pT292 reduced | Functionally significant mutation disrupting feedback |
MAP2K1/2 mutation (+) / pT292 normal | Mutation not affecting feedback regulation |
MAP2K1/2 wild-type / pT292 reduced | Alternative pathway dysregulation |
MAP2K1/2 wild-type / pT292 normal | Intact pathway regulation |
Therapeutic Decision Support:
For targeted therapies: Patients with impaired T292 phosphorylation (disrupted feedback) may benefit from combination treatments that address pathway reactivation
For immunotherapy: Based on study data, MAP2K1/2 mutations showed:
Research Implementation Strategy:
Develop standardized immunohistochemistry protocols for detecting T292 phosphorylation in FFPE samples
Conduct retrospective analysis of T292 phosphorylation in patient cohorts with known treatment outcomes
Include T292 phosphorylation analysis in prospective clinical trials as an exploratory endpoint
Correlate with other established biomarkers (e.g., TMB, PD-L1 expression)
Future prospective studies should validate the predictive value of combined MAP2K1/2 mutation status and T292 phosphorylation levels to develop clinically applicable stratification algorithms .
Several emerging technologies hold promise for advancing the detection and functional analysis of MAP2K1 T292 phosphorylation with improved sensitivity, specificity, and spatiotemporal resolution:
Mass Spectrometry-Based Approaches:
Targeted Phosphoproteomics: Selected Reaction Monitoring (SRM) or Parallel Reaction Monitoring (PRM) can quantify T292 phosphopeptides with high sensitivity and specificity
Absolute Quantification: Using isotopically labeled synthetic phosphopeptides as internal standards enables absolute quantification of phosphorylation stoichiometry
Single-Cell Phosphoproteomics: Emerging techniques for analyzing phosphorylation events at single-cell resolution can reveal heterogeneity in MAP2K1 regulation across cell populations
Advanced Imaging Technologies:
Super-Resolution Microscopy: Techniques like STORM or PALM combined with phospho-specific antibodies can reveal subcellular localization of phosphorylated MAP2K1
Förster Resonance Energy Transfer (FRET): Genetically encoded FRET biosensors for MAP2K1 can monitor T292 phosphorylation dynamics in living cells in real-time
Spatial Transcriptomics Integration: Combining phospho-protein detection with spatial transcriptomics can link T292 phosphorylation to local gene expression patterns
Functional Genomics Approaches:
CRISPR Base Editing: Precise modification of the T292 site to non-phosphorylatable residues without disrupting protein expression
Phospho-mimetic Mutations: Expression of T292D/E mutants to model constitutive phosphorylation
Optogenetic Control: Light-inducible ERK activation systems to study temporal dynamics of T292 phosphorylation
Microfluidic and Lab-on-a-Chip Systems:
Phospho-Flow-Seq: Combining phospho-flow cytometry with single-cell RNA sequencing
Microfluidic Western Blotting: Miniaturized systems requiring minimal sample input (valuable for limited clinical specimens)
Organ-on-a-Chip Models: Testing pathway dynamics in physiologically relevant microenvironments
Computational Approaches:
Deep Learning Image Analysis: AI-assisted quantification of phospho-T292 immunohistochemistry in tissue samples
Dynamic Pathway Modeling: Incorporating T292 phosphorylation kinetics into mathematical models of MAPK pathway behavior
Multi-omics Data Integration: Correlating phosphorylation data with transcriptomics, metabolomics and clinical outcomes
These technologies could significantly advance our understanding of MAP2K1 T292 phosphorylation beyond current antibody-based methods, enabling more sophisticated analyses of its role in normal physiology and disease states .
Targeting the regulatory mechanisms involving MAP2K1 T292 phosphorylation represents an innovative approach to cancer treatment that could address limitations of current MAPK pathway inhibitors:
Disrupting Negative Feedback for Enhanced Therapy:
Rationale: T292 phosphorylation mediates negative feedback that limits MAPK pathway activation. In certain contexts, enhancing this feedback could suppress oncogenic signaling.
Approach: Development of small molecules that mimic or enhance ERK-mediated phosphorylation of T292, potentially stabilizing the phosphorylated state
Potential Application: Tumors with hyperactivated MAPK signaling but intact feedback machinery could be sensitive to enhanced negative regulation
Targeting Feedback-Escape Mechanisms:
Rationale: Some cancers develop resistance by evading T292-mediated feedback inhibition
Approaches:
Synergistic Combination Strategies:
With MEK Inhibitors: Compounds modulating T292 regulatory mechanisms could enhance durability of response to traditional MEK inhibitors
With Immunotherapies: Based on data showing potential correlation between MAP2K1/2 mutations and immunotherapy response, targeting T292 regulatory mechanisms might sensitize tumors to immune checkpoint inhibitors
Proposed Combination Strategy:
Therapeutic Component | Target | Mechanism | Expected Outcome |
---|---|---|---|
MEK inhibitor | MEK catalytic activity | Block downstream signaling | Initial pathway inhibition |
T292 regulatory modulator | Feedback mechanism | Prevent pathway reactivation | Prevent resistance development |
Immunotherapy (CTLA-4 inhibitor) | Immune checkpoint | Enhance anti-tumor immunity | Sustained tumor control |
Patient Selection Strategies:
Therapeutic Monitoring Approach:
This targeted approach to pathway regulation, rather than simple inhibition, represents a paradigm shift in MAPK pathway-directed therapies that could potentially overcome current limitations in treatment durability and response .