The antibody targets the phosphorylated Y307 residue of PPP2CA, a key modification that inactivates PP2A’s phosphatase activity . PP2A is a tumor suppressor implicated in dephosphorylating proteins involved in cell cycle regulation, apoptosis, and DNA repair . Phosphorylation at Y307 disrupts PP2A’s function, contributing to oncogenic processes .
This antibody is produced via recombinant DNA technology, ensuring batch consistency and specificity:
Immunization: Animals are immunized with a synthetic phospho-peptide to generate B cells producing anti-phospho-Y307 antibodies .
Cloning: Positive B cells are isolated, and single clones (e.g., 3F11) are identified .
Expression: Heavy and light chains are amplified via PCR, cloned into plasmids, and transfected into host cells (e.g., HEK293F) for antibody production .
Purification: Affinity chromatography isolates the antibody from cell culture supernatant .
The antibody detects phosphorylated PPP2CA in human cell lines:
Cell Line | Treatment | Observed Signal | Source |
---|---|---|---|
A549 | EGF-treated | Strong 35 kDa band | |
A549 | Untreated | Basal phosphorylation | |
HEK293T | Native | Detectable PPP2CA-Y307 | |
Hela | Native | Positive signal |
Phosphorylation at Y307 is linked to PP2A inactivation in cancers, including metastatic colorectal cancer (CRC) . Studies suggest:
Clinical Relevance: p-PPP2CA may predict poor outcomes in CRC and guide PP2A-activating therapies .
Mechanistic Insight: Y307 phosphorylation disrupts PP2A’s interaction with regulatory subunits, impairing its tumor-suppressive functions .
Sample | Antibody Concentration | Secondary Antibody | Signal Quality |
---|---|---|---|
A549 (EGF-treated) | 0.95 μg/mL | Goat anti-rabbit IgG (1:50,000) | Strong 35 kDa band |
A549 (untreated) | 0.95 μg/mL | Goat anti-rabbit IgG (1:50,000) | Basal phosphorylation |
HEK293T | 1:500–1:1,000 | N/A | Detectable PPP2CA-Y307 |
Specificity: No cross-reactivity reported for non-phosphorylated PPP2CA .
Sensitivity: Detects endogenous phosphorylation in untreated cells .
Cancer: Y307 phosphorylation correlates with metastasis and therapy resistance .
Neurodegeneration: PP2A regulates tau phosphorylation, linking to Alzheimer’s disease .
Immune Regulation: Modulates T-cell activation and cytokine signaling .
The phospho-PPP2CA (Y307) recombinant monoclonal antibody is produced using protein technology and DNA recombinant techniques. Initially, animals are immunized with a synthetic peptide derived from human phospho-PPP2CA (Y307), leading to the generation of B cells. These B cells are then carefully screened to isolate positive clones, followed by single clone identification. The light and heavy chains of the phospho-PPP2CA (Y307) antibody are amplified via PCR and integrated into a plasmid vector to construct a recombinant vector. This recombinant vector is subsequently transfected into host cells to facilitate antibody expression. The phospho-PPP2CA (Y307) recombinant monoclonal antibody is purified from the cell culture supernatant using affinity chromatography. Rigorous validation procedures are conducted to ensure its accuracy and efficacy for ELISA and WB applications. The phospho-PPP2CA (Y307) recombinant monoclonal antibody serves as a valuable tool for detecting human phospho-PPP2CA (Y307) protein in research settings.
PP2A is the primary phosphatase for microtubule-associated proteins (MAPs). Its activity modulates the function of phosphorylase B kinase, casein kinase 2, mitogen-stimulated S6 kinase, and MAP-2 kinase. In oocytes, PP2A collaborates with SGO2 to safeguard centromeric cohesin from separase-mediated cleavage specifically during meiosis I. It can dephosphorylate SV40 large T antigen and p53/TP53. PP2A activates RAF1 by dephosphorylating it at 'Ser-259'. It mediates dephosphorylation of WEE1, preventing its ubiquitin-mediated proteolysis, leading to increased WEE1 protein levels and promoting the G2/M checkpoint. PP2A mediates dephosphorylation of MYC, promoting its ubiquitin-mediated proteolysis; its interaction with AMBRA1 enhances interaction between PPP2CA and MYC. It also mediates dephosphorylation of FOXO3, promoting its stabilization; interaction with AMBRA1 enhances interaction between PPP2CA and FOXO3.
Phospho-PPP2CA (Y307) Recombinant Monoclonal Antibody [3F11] is a primary antibody designed to recognize the phosphorylated tyrosine 307 residue of the catalytic subunit alpha isoform of protein phosphatase 2A (PPP2CA). The antibody is a recombinant monoclonal antibody produced in HEK293F cells with rabbit IgG isotype. It is unconjugated (not labeled with any tags or fluorophores) and reacts specifically with human samples .
The key specifications of this antibody are summarized in the following table:
Parameter | Specification |
---|---|
Type | Primary Antibody |
Clonality | Monoclonal |
Clone Name | 3F11 |
Host | HEK293F Cell |
Reactivity | Human |
Isotype | Rabbit IgG |
Label | Unconjugated |
Applications | ELISA, Western Blot (WB) |
Recommended Dilution for WB | 1:500-1:5000 |
Immunogen | Synthesized peptide derived from human Phospho-PPP2CA (Y307) |
Purification Method | Affinity Chromatography |
Protein Phosphatase 2A (PP2A) is a major serine/threonine phosphatase involved in regulating numerous enzymes, signal transduction pathways, and cellular processes. PPP2CA is the catalytic subunit alpha isoform of PP2A.
PP2A's functions include:
Serving as the major phosphatase for microtubule-associated proteins (MAPs)
Modulating the activity of multiple kinases including phosphorylase B kinase, casein kinase 2, mitogen-stimulated S6 kinase, and MAP-2 kinase
Cooperating with SGO2 to protect centromeric cohesin during meiosis I
Mediating dephosphorylation of WEE1, preventing its ubiquitin-mediated proteolysis and promoting the G2/M checkpoint
Mediating dephosphorylation of MYC and FOXO3, promoting their ubiquitin-mediated proteolysis or stabilization
Catalyzing dephosphorylation of NLRP3's pyrin domain, promoting inflammasome assembly
Participating in striatin-interacting phosphatase and kinase (STRIPAK) complexes, which regulate multiple signaling pathways including Hippo, MAPK, nuclear receptor and cytoskeleton remodeling
To optimize Western blot protocols with Phospho-PPP2CA (Y307) antibody:
Sample preparation: Include phosphatase inhibitors (e.g., sodium orthovanadate, sodium fluoride) in lysis buffers to preserve phosphorylation status.
Blocking: Use 5% BSA in TBST rather than milk, as milk contains phosphoproteins that may interfere with phospho-specific antibody binding.
Antibody dilution: Start with a 1:1000 dilution within the recommended range (1:500-1:5000) . Optimize based on signal-to-noise ratio.
Incubation time: Incubate with primary antibody overnight at 4°C to maximize specific binding.
Controls:
Signal detection: Use enhanced chemiluminescence (ECL) or fluorescence-based detection systems.
Validation: Confirm specificity by using peptide competition assays or comparing with alternative detection methods.
When studying PP2A phosphorylation at Y307, the following controls are essential:
Phosphatase treatment control: Treat a portion of your sample with lambda phosphatase to confirm the antibody is detecting phosphorylated protein.
Stimulus controls: Include samples with known modulators of PP2A phosphorylation:
Mutant controls: Use PP2A constructs with site-directed mutations:
Immunoprecipitation controls: Validate phosphorylation using:
Anti-phosphotyrosine antibodies after PP2A immunoprecipitation
Reciprocal immunoprecipitation with anti-phosphotyrosine followed by PP2A detection
Alternative methods: Confirm findings using:
Mass spectrometry to directly identify phosphorylation sites
Phos-tag gels to separate phosphorylated from non-phosphorylated forms
These controls are particularly important given the contradictory findings about Y307 phosphorylation discussed in section 3.
Recent research significantly challenges the long-held dogma that Y307 is the primary phosphorylation site of PP2A catalytic subunit (PP2Ac), raising important considerations for researchers:
Mass spectrometry (MS) studies of Src-transformed mouse embryo fibroblasts (MEFs) have identified endogenous PP2Ac phosphorylation at Y127 and Y284, but not at Y307 . Multiple discovery-mode MS investigations across various oncogenic/transformed cells and human cancers have similarly identified Y284 phosphorylation, but not Y307 .
Even in targeted MS studies using HEK293T cells overexpressing both constitutively active Src and PP2Ac, Y307 phosphorylation was detected only at very low levels . Experiments using Y307F point mutants of PP2Ac revealed very similar levels of tyrosine phosphorylation compared to wild-type PP2Ac in response to Src activation or pervanadate treatment .
These findings suggest that:
Y307 is not the predominant tyrosine phosphorylation site of PP2Ac
Y127 and Y284 appear to be the major sites targeted by Src family kinases (SFKs)
Most studies assessing PP2A Y307 phosphorylation have relied on antibodies that may lack specificity
This represents a paradigm shift in our understanding of PP2A regulation and raises questions about the utility and specificity of many commercially available "anti-pY307 PP2Ac" antibodies.
The comparative significance of these phosphorylation sites reveals a complex regulatory mechanism:
These phosphorylation events enhance interaction between PP2Ac and SFKs
Y284 phosphorylation particularly promotes dissociation of the regulatory Bα subunit, altering PP2A substrate specificity
Mutation of these sites (Y127/284F and Y284F) prevents SFK-mediated phosphorylation of Tau at pSer202 (CP13 epitope), a pathological hallmark of Alzheimer's disease
These mutations also prevent SFK-dependent activation of ERK, a major growth regulatory kinase upregulated in many cancers
Previously believed to be the sole site of phosphorylation on PP2Ac
Traditionally thought to be essential for catalytic inactivation of PP2A and v-Src-mediated cell transformation
Recent evidence suggests this site is phosphorylated at very low levels, if at all, under physiological conditions
Many commercially available "anti-pY307" antibodies have been shown to recognize both wild-type and Y307F mutant PP2Ac equally, raising questions about their specificity
The "Goldilocks phenomenon" refers to the observation that PPP2CA exhibits concentration-dependent opposing functions in cancer cells - specifically in neuroblastoma (NB). This phenomenon has significant implications for research:
At low expression levels, PPP2CA functions as an essential survival gene for cancer cells
At high expression levels, PPP2CA acts as a tumor suppressor
Reduction of PPP2CA by knock-down decreased growth of neuroblastoma cells
Complete ablation of PPP2CA by knock-out was not tolerated by these cells
Neuroblastoma cells show addiction to PPP2CA, which is augmented by MYCN activation
SET, an endogenous inhibitor of PP2A, was overexpressed in poor-prognosis neuroblastoma
The SET inhibitor OP449 effectively decreased viability of neuroblastoma cells, consistent with a tumor suppressor function of PPP2CA when its activity is enhanced
Researchers must consider the baseline expression level of PPP2CA when interpreting experimental results
Therapeutic strategies targeting PPP2CA must account for this dual functionality
Simple activation or inhibition of PPP2CA may produce contradictory effects depending on cellular context
Combined approaches that maintain PPP2CA within an optimal functional range may be necessary
This phenomenon explains why both PP2A inhibitors and activators have shown anti-cancer effects in different contexts and suggests that precise modulation, rather than simple activation or inhibition, may be required for therapeutic applications.
Given recent findings questioning the specificity of commercial pY307 antibodies , researchers should implement rigorous validation protocols:
Genetic validation:
Express wild-type PP2A alongside Y307F mutant in a cellular system
If the antibody is specific for pY307, it should not recognize the Y307F mutant by Western blot or immunofluorescence
Note: Some commercial "anti-pY307" antibodies have been shown to recognize both wild-type and Y307F mutants equally
Phosphatase treatment controls:
Treat cell lysates with lambda phosphatase and confirm loss of antibody recognition
Include both positive controls (untreated) and negative controls (phosphatase-treated)
Peptide competition assays:
Pre-incubate antibody with phospho-Y307 peptide and non-phosphorylated control peptide
Specific binding should be blocked by phospho-peptide but not by non-phospho-peptide
Mass spectrometry validation:
Perform targeted mass spectrometry to directly assess Y307 phosphorylation status
Compare MS results with antibody-based detection methods
Multiple antibody approach:
Use antibodies from different vendors or clones
Compare detection patterns to identify potential non-specific binding
Kinase manipulation:
Modulate SFK activity using inhibitors (PP2) or activators
Assess correlation between kinase activity and antibody signal
Cross-reactivity assessment:
Test the antibody against Y127F, Y284F, and Y127/284F mutants
Evaluate whether the antibody cross-reacts with other phosphotyrosine sites
To address contradictions between commercial antibody specifications and recent research findings , researchers should employ these methodological approaches:
Integrated multi-method detection strategy:
Combine antibody-based methods with mass spectrometry
Use Phos-tag SDS-PAGE to separate and identify phosphorylated species
Apply proximity ligation assays to detect and localize phosphorylated PP2A
Site-directed mutagenesis comparative analysis:
Phospho-specific antibody generation and validation:
Develop new antibodies specifically validated against mutant controls
Characterize antibodies using multiple cell types and treatments
Confirm specificity using peptide competition and phosphatase treatment
Functional correlation analysis:
Compare PP2A activity measurements with phosphorylation status detected by various methods
Assess correlation between detected phosphorylation and functional outcomes like regulatory subunit binding
Quantitative phosphoproteomics:
Time-course and stimulus-response analysis:
Phospho-PPP2CA antibodies can provide valuable insights into neurodegenerative disease mechanisms through carefully designed experimental approaches:
Tau phosphorylation studies in Alzheimer's disease (AD):
Recent research has shown that SFK-mediated phosphorylation of PP2Ac at Y284 alters substrate specificity and influences Tau phosphorylation at the CP13 (pSer202) epitope, a pathological hallmark of AD
Researchers can use phospho-specific antibodies alongside PP2A mutants (Y127F, Y284F) to investigate how PP2A tyrosine phosphorylation impacts Tau hyperphosphorylation
Comparative immunohistochemistry of phospho-PP2A and phospho-Tau in AD brain sections can reveal spatial relationships
PP2A-SFK interaction analysis:
Phospho-PP2A antibodies can be used in co-immunoprecipitation studies to assess how PP2A phosphorylation affects its interaction with Src family kinases in neuronal models
Proximity ligation assays can visualize these interactions in situ in brain tissue
Regulatory subunit displacement mechanism:
In vivo phosphorylation monitoring:
Tracking PP2A phosphorylation status in animal models of neurodegeneration at different disease stages
Correlating changes in PP2A phosphorylation with cognitive deficits and neuropathology
Therapeutic intervention assessment:
Evaluating how potential therapeutic compounds affect PP2A phosphorylation status and downstream substrate phosphorylation
Using phospho-PP2A antibodies as pharmacodynamic markers in preclinical studies
The newly discovered SFK-mediated PP2A regulatory mechanism has profound implications for cancer research, challenging existing paradigms and opening new therapeutic avenues:
Revised understanding of PP2A regulation in cancer:
The finding that SFKs primarily phosphorylate PP2A at Y127 and Y284 rather than Y307 fundamentally changes our understanding of how oncogenic kinases regulate this crucial phosphatase
Rather than simply inhibiting catalytic activity, SFK-mediated phosphorylation appears to alter PP2A substrate specificity by modulating regulatory subunit binding
Impact on SFK-ERK signaling axis:
Dual role of PP2A in cancer (Goldilocks phenomenon):
PP2A addiction in cancer cells:
SET inhibition as a therapeutic strategy:
Biomarker potential:
The phosphorylation status of PP2A at Y127 and Y284 could serve as biomarkers for SFK activity and potential response to SFK inhibitors in cancer
Monitoring these sites might better predict therapeutic outcomes than the previously used Y307 phosphorylation
Preserving PP2A phosphorylation status requires specific sample preparation considerations:
Lysis buffer optimization:
Use buffers containing phosphatase inhibitors: sodium orthovanadate (1-2 mM), sodium fluoride (10 mM), β-glycerophosphate (10 mM), and sodium pyrophosphate (5 mM)
Include protease inhibitors to prevent degradation
Maintain cold temperature throughout processing (4°C)
Consider using commercial phospho-protein preservation buffers
Cell harvesting techniques:
Direct lysis in the culture dish is preferred over trypsinization
If cell scraping is necessary, perform quickly and transfer immediately to cold lysis buffer
For tissues, snap-freeze in liquid nitrogen immediately after collection
Denaturing conditions:
Include sufficient detergent (1% NP-40 or 0.5% Triton X-100) to solubilize membrane-associated PP2A
Add SDS (0.1%) to denature phosphatases that might dephosphorylate during processing
Heat samples at 95°C for 5 minutes in Laemmli buffer containing 5% β-mercaptoethanol
Phosphatase treatment controls:
Process parallel samples with and without lambda phosphatase treatment
This provides a negative control for phospho-specific antibody detection
Stabilization strategies:
Fractionation considerations:
If performing subcellular fractionation, maintain phosphatase inhibitors in all buffers
Validate phosphorylation status in each fraction with appropriate controls
Storage conditions:
Store lysates at -80°C with phosphatase inhibitors
Avoid multiple freeze-thaw cycles
Add glycerol (10%) for cryoprotection if multiple uses are anticipated
Determining optimal experimental conditions for studying PP2A phosphorylation requires systematic optimization:
Cell model selection:
For SFK-mediated phosphorylation at Y127/Y284, use:
For potential Y307 phosphorylation, use:
Stimulus optimization:
Time-course analysis:
Monitor phosphorylation at different time points (5, 15, 30, 60 minutes) after stimulation
For EGF stimulation, focus on early time points (5-15 minutes)
For constitutively active kinase expression, 24-48 hours post-transfection is typically optimal
Detection method selection:
Western blotting: Optimize antibody dilution (start with 1:1000) and incubation conditions
Immunoprecipitation: Use either PP2A antibodies followed by phosphotyrosine detection or vice versa
Phos-tag SDS-PAGE: Optimize acrylamide percentage (6-8%) and Phos-tag concentration (25-100 μM)
Mass spectrometry: Consider SILAC or TMT labeling with phosphotyrosine enrichment
Mutant analysis system:
Comparative kinase analysis:
This systematic approach will help researchers determine the optimal conditions for studying each phosphorylation site while avoiding potential pitfalls based on outdated models of PP2A regulation.
Several emerging technologies hold promise for advancing our understanding of PP2A phosphorylation dynamics:
Genetically encoded phosphorylation sensors:
FRET-based sensors specifically designed for PP2A phosphorylation sites
These would allow real-time monitoring of phosphorylation/dephosphorylation events in living cells
Could reveal compartment-specific dynamics and temporal regulation
CRISPR-based phosphorylation site editing:
Precise genome editing to mutate endogenous PP2A phosphorylation sites (Y127, Y284, Y307)
Avoids overexpression artifacts associated with transfection-based approaches
Can be combined with conditional systems to study tissue-specific effects
Proximity-dependent labeling technologies:
BioID or TurboID fused to PP2A to identify proximity interactors dependent on phosphorylation status
APEX2 labeling to map the spatiotemporal organization of phosphorylated PP2A pools
Would reveal how phosphorylation affects PP2A interaction networks
Single-molecule tracking:
Tracking individual PP2A molecules in living cells using quantum dots or photoactivatable fluorophores
Could reveal how phosphorylation affects PP2A diffusion, localization, and complex formation
May uncover heterogeneity in PP2A populations that bulk analyses miss
Advanced mass spectrometry techniques:
Targeted parallel reaction monitoring (PRM) for absolute quantification of site-specific phosphorylation
Crosslinking mass spectrometry to capture phosphorylation-dependent structural changes
Top-down proteomics to analyze intact PP2A complexes with multiple modifications
Optogenetic control of kinase activity:
Light-controlled activation of SFKs to achieve temporal precision in PP2A phosphorylation
Would enable study of rapid phosphorylation dynamics without chemical perturbations
Could be combined with live-cell imaging to correlate phosphorylation with functional outcomes
Cryo-EM structural analysis:
High-resolution structures of phosphorylated versus non-phosphorylated PP2A complexes
Would reveal how phosphorylation at different sites affects holoenzyme assembly
Could guide structure-based drug design for phosphorylation-specific modulators
The revised understanding of PP2A phosphorylation offers several promising avenues for therapeutic development:
Site-specific PP2A modulators:
Rather than general PP2A activators or inhibitors, develop compounds that selectively interfere with Y127 or Y284 phosphorylation
These could modulate PP2A substrate specificity without completely inhibiting catalytic activity
Potential to disrupt specific pathological PP2A functions while preserving essential ones
Targeted degradation approaches:
SFK-PP2A interface disruptors:
Design peptides or small molecules that specifically disrupt the interaction between SFKs and PP2A
This would prevent phosphorylation while potentially preserving PP2A activity
Could selectively block pathological SFK-mediated PP2A regulation
Regulatory subunit-focused strategies:
Dual-targeting approaches:
Combine SFK inhibitors with SET inhibitors to simultaneously reduce aberrant PP2A phosphorylation and enhance PP2A activity
This two-pronged approach could be particularly effective in cancers with high SFK activity and SET expression
Would address both catalytic inhibition and substrate redirection mechanisms
Biomarker-guided therapies:
Use PP2A phosphorylation status (Y127/Y284 vs. Y307) as biomarkers to guide therapeutic decisions
Select patients most likely to benefit from SFK inhibitors or PP2A modulators based on phosphorylation profile
Monitor treatment efficacy through changes in phosphorylation status
Stabilization of regulatory interactions: