PP2AA1 Antibody specifically recognizes the regulatory Aα subunit (PPP2R1A) of PP2A, a heterotrimeric enzyme composed of:
Scaffolding subunit (Aα/β): Structural backbone for holoenzyme assembly .
Regulatory subunit (B): Determines substrate specificity and localization .
PP2A accounts for 90% of cellular serine/threonine phosphatase activity , making PP2AA1 a pivotal tool for studying PP2A-mediated signaling in cancer, neurodegeneration, and immunity .
PP2AA1 maintains PP2A stability and recruits regulatory subunits to modulate pathways like PI3K/AKT, MAPK, and NF-κB . Dysregulation of PP2A is linked to Alzheimer’s disease, cancer, and autoimmune disorders .
PP2AA1 antibodies are validated for:
Western Blot (WB): Detects endogenous PP2A at ~65 kDa in human, mouse, and rat lysates .
Immunohistochemistry (IHC): Localizes PP2A in paraffin-embedded tissues .
Immunoprecipitation (IP): Identifies PP2A interactomes (e.g., Dab2 in thrombosis studies) .
Functional Studies: Elucidates PP2A’s role in T-cell differentiation, B-cell responses, and endothelial signaling .
Cancer Immunotherapy: PP2A inhibitors (e.g., LB-100) combined with checkpoint blockers show efficacy in glioma and melanoma .
Autoimmunity: PP2AA1-associated signaling in T-cells drives SLE and rheumatoid arthritis via Th17 differentiation .
Neurological Disorders: PP2A activators (tricyclic sulfonamides) reduce Aβ and tau aggregation in Alzheimer’s models .
Specificity Controls: Preabsorption with phospho/dephospho-peptides or phosphatase treatment validates epitope recognition .
Activity Assays: PP2A immunoprecipitated with PP2AA1 antibodies shows altered phosphatase activity in thrombosis and cancer models .
The A subunit of protein phosphatase 2A (PP2A) acts as a scaffolding protein, crucial for assembling the catalytic subunit and a variable regulatory B subunit. It functions as a positive regulator of PP2A catalytic activity, conferring resistance to inhibitors such as okadaic acid and cantharidin. PP2AA1 is implicated in various developmental processes, including seedling and floral development, root gravitropism, and stomatal opening regulation. Furthermore, it plays a significant role in auxin efflux regulation, particularly during basipetal (tip-to-base) auxin transport in roots. Its involvement extends to the perception of auxin efflux inhibitors like 1-N-naphthylphthalamic acid (NPA) and semicarbazone I (SCB-I). PP2AA1 modulates the ethylene response in hypocotyls and stems and acts as a positive transducer of early abscisic acid (ABA) signaling. The PP2AA1-PP2A4-B'ZETA/B'ETA holoenzyme complex functions as a negative regulator of plant innate immunity by controlling the phosphorylation state and activation of BAK1 within surface-localized immune receptor complexes.
Further research highlights the multifaceted roles of PP2AA1:
PP2AA1 (also referred to as PPP2CA) is the catalytic subunit alpha isoform of Protein Phosphatase 2A (PP2A), a major serine/threonine phosphatase that plays crucial roles in numerous cellular processes. PP2A functions as a heterotrimer consisting of a catalytic subunit (C), a structural subunit (A), and a variable regulatory subunit (B). The PP2AA1 catalytic subunit is involved in regulating:
Multiple signaling pathways including Hippo, MAPK, and nuclear receptor pathways
Cell cycle progression and G2/M checkpoint through WEE1 dephosphorylation
Oncogene regulation through dephosphorylation of proteins like MYC and FOXO3
Inflammasome assembly via NLRP3 pyrin domain dephosphorylation
These diverse functions make PP2AA1 a critical target for investigation in various biological contexts, from normal cellular homeostasis to disease states.
Antibody selection should be based on:
Application compatibility: Different antibodies are optimized for specific applications such as Western blotting (WB), immunoprecipitation (IP), immunohistochemistry (IHC), or immunofluorescence (IF). For example, the PP2A-Aα/β Antibody (B-1) is validated for WB, IP, IF, and ELISA applications .
Species reactivity: Verify that the antibody has been validated in your experimental species. The PP2A-alpha antibody from Abcam (ab137825) reacts with human and mouse samples , while the PP2A-Aα/β Antibody (B-1) can detect PP2A-Aα/β in mouse, rat, and human samples .
Epitope recognition: Consider whether you need an antibody targeting a specific region or post-translational modification of PP2AA1. For example, ab137825 recognizes an epitope within amino acids 1-250 of human PPP2CA .
Validation evidence: Review provided validation data including western blots showing predicted band sizes (35 kDa for PP2AA1) and examine citation records for evidence of successful application in published research.
Clonality considerations: Monoclonal antibodies offer high specificity for single epitopes, while polyclonal antibodies may provide higher sensitivity by recognizing multiple epitopes.
Several experimental artifacts can compromise research with PP2AA1 antibodies:
Cross-reactivity with unmodified forms: Some antibodies marketed as specific for post-translationally modified forms (e.g., phospho-Tyr307 PP2A antibodies) have been shown to also detect unmodified PP2AA1 .
Sensitivity to neighboring modifications: Antibody binding may be affected by modifications on residues near the target epitope. For example, the E155 and F-8 clones show reduced binding to PP2AA1 when Leu309 is methylated .
Non-specific bands: Particularly in complex lysates, antibodies may bind to proteins of similar molecular weight to PP2AA1 (35 kDa).
Batch-to-batch variation: Especially with polyclonal antibodies, different production lots may show variable specificity and sensitivity.
Fixation artifacts: For immunohistochemistry and immunofluorescence applications, different fixation methods can affect epitope accessibility and antibody binding.
To minimize these artifacts, proper controls including knockdown/knockout samples are essential for validating antibody specificity in your experimental system.
Distinguishing between PP2AA1 phosphorylation states requires careful consideration of antibody specificity:
Recent research has revealed significant limitations in antibodies previously believed to be phospho-specific. For example, extensive validation of commercially available phospho-Tyr307 PP2AC antibodies (including clones E155, F-8, and antibodies from R&D Systems) demonstrated that these antibodies cannot reliably differentiate between phosphorylated and unphosphorylated forms of PP2AA1 .
For accurate detection of phosphorylation states:
Use alternative methods: Combine antibody-based detection with mass spectrometry to verify phosphorylation status.
Implement appropriate controls: Include phosphatase treatment of samples to remove phosphorylation and compare signal intensity.
Phospho-mimetic mutants: Generate Y307E or Y307D phospho-mimetic mutants and Y307F phospho-null mutants as controls for antibody validation.
Consider neighboring modifications: Be aware that phosphorylation at Thr304 and methylation at Leu309 can affect antibody binding, potentially confounding interpretation of phosphorylation status at Tyr307 .
Comprehensive validation requires multiple approaches:
Genetic knockout/knockdown controls:
siRNA/shRNA knockdown of PP2AA1
CRISPR/Cas9 knockout cell lines
Comparing signal in wild-type vs. knockout samples by Western blot
Mutant expression systems:
Peptide competition assays:
Pre-incubate antibody with blocking peptides corresponding to the target epitope
Observe disappearance of specific signal
Multiple antibody validation:
Use different antibodies recognizing distinct epitopes
Compare patterns of detection across applications
Post-translational modification (PTM) manipulation:
Treat samples with phosphatases to remove phosphorylation
Expose cells to stimuli known to induce specific modifications (e.g., EGF treatment for tyrosine phosphorylation)
For example, researchers demonstrated that the E155 and F-8 antibodies detect both wild-type and Y307F mutant forms of PP2AA1 with equal intensity, revealing they cannot distinguish phosphorylated from unphosphorylated forms at Tyr307 .
Assessing PP2AA1 activity requires methods beyond simple antibody detection:
PP2A activity assays:
Immunoprecipitate PP2A complexes using antibodies against the catalytic or structural subunits
Measure phosphatase activity using synthetic phosphopeptide substrates
Quantify released phosphate using malachite green or similar colorimetric methods
Endogenous substrate phosphorylation:
Pharmacological approaches:
Use specific PP2A inhibitors (e.g., okadaic acid at appropriate concentrations)
Compare substrate phosphorylation with and without inhibitors
Separation of PP2A complexes:
Employ immunoprecipitation to isolate specific PP2A holoenzymes
Assess activity of distinct PP2A complexes containing different regulatory subunits
In one study, researchers successfully measured PP2A activity in tolerized peritoneal macrophages, correlating increased PP2A activity with decreased TNF-α secretion upon second LPS stimulation , demonstrating the feasibility of activity measurements in complex cellular contexts.
For successful immunofluorescence with PP2AA1 antibodies:
Fixation optimization:
Antibody dilution and incubation:
Controls and counterstaining:
Signal amplification and detection:
For weak signals, consider tyramide signal amplification
Use confocal microscopy for optimal subcellular localization
Image acquisition settings:
Adjust exposure to avoid signal saturation
Maintain consistent settings between experimental and control samples
For example, successful visualization of PP2AA1 in HeLa cells has been achieved using methanol fixation with ab137825 at 1/500 dilution, counterstained with alpha-tubulin .
Optimization strategies for Western blotting include:
Sample preparation:
Gel selection and transfer conditions:
10-12% SDS-PAGE gels are appropriate for resolving the 35 kDa PP2AA1
Semi-dry or wet transfer systems work well (20-30V overnight or 100V for 1 hour)
Use PVDF membranes for optimal protein binding
Blocking and antibody incubation:
Detection and quantification:
Use HRP-conjugated secondary antibodies with ECL detection
For quantification, ensure exposure times avoid signal saturation
Normalize to appropriate loading controls (e.g., GAPDH, β-actin)
Stripping and reprobing:
Gentle stripping buffers can allow sequential probing for phosphorylated and total protein
Validate complete stripping before reprobing
The expected band size for PP2AA1 is 35 kDa, and this should be verified using appropriate molecular weight markers .
Essential controls for immunoprecipitation experiments include:
Input control:
Always run an aliquot of starting material (5-10% of IP amount)
Use for comparison to immunoprecipitated material
Negative antibody control:
Beads-only control:
Incubate lysate with capture matrix without antibody
Identifies proteins binding non-specifically to beads
Competitive blocking:
Reciprocal IP:
For protein-protein interaction studies, perform IP with antibodies against each partner
Confirms interactions from both perspectives
Validation in knockdown/knockout samples:
Perform IP in cells where PP2AA1 expression is reduced/eliminated
Demonstrates specificity of immunoprecipitation
For PP2AA1 complex studies, consider using alternative antibodies against different PP2A subunits (A, B, or C) to validate interactions and complex formation.
When faced with conflicting results:
Epitope differences:
Clonality considerations:
Monoclonal antibodies target single epitopes, while polyclonal antibodies recognize multiple epitopes
Polyclonal antibodies may maintain signal despite modification of individual epitopes
Sensitivity to neighboring modifications:
Validation approach:
Create a validation matrix with multiple antibodies across different applications
Include knockout/knockdown controls for each antibody
Use orthogonal methods (mass spectrometry) to confirm antibody results
Resolution strategies:
When antibodies disagree, trust results from antibodies with most extensive validation
For critical findings, confirm with non-antibody-based methods
Consider that both results may be correct if detecting different subpopulations of PP2AA1
The study by Frohner et al. (2020) highlights how reinterpretation of previous work may be necessary when antibody specificity is thoroughly examined .
The discovery that widely used phospho-Tyr307 antibodies cannot differentiate between phosphorylated and unphosphorylated forms has profound implications:
This case highlights the critical importance of rigorous antibody validation and demonstrates how insufficient validation can lead an entire field in potentially misleading directions .
A comprehensive experimental design would include:
Multiple detection approaches:
Antibody-based detection with extensively validated antibodies
Mass spectrometry analysis of immunoprecipitated PP2AA1
Phospho-protein staining (Pro-Q Diamond) followed by total protein staining
Genetic manipulation:
Expression of tagged wild-type PP2AA1 alongside phospho-null (Y307F) and phospho-mimetic (Y307E) mutants
CRISPR-mediated genomic editing to create endogenous Y307F mutations
Pharmacological interventions:
Treatment with phosphatase inhibitors (okadaic acid, calyculin A)
Tyrosine kinase inhibitors targeting kinases implicated in Y307 phosphorylation
Phosphatase treatment of cellular extracts
Functional correlation:
Parallel measurement of PP2A activity using phosphatase assays
Correlation of activity with phosphorylation status
Assessment of downstream substrate phosphorylation
Temporal dynamics:
Time-course experiments following stimulation
Correlation of changes in phosphorylation with functional outcomes
This multi-faceted approach would minimize reliance on any single detection method and provide multiple lines of evidence regarding PP2AA1 phosphorylation status and its functional significance.
Advanced proteomics offers powerful solutions for PP2AA1 research:
Parallel Reaction Monitoring (PRM):
Targeted mass spectrometry approach for precise quantification of specific PP2AA1 phosphopeptides
Can distinguish between different phosphorylated forms with site-specific resolution
Provides absolute quantification of modification stoichiometry
Crosslinking Mass Spectrometry (XL-MS):
Maps interaction interfaces between PP2AA1 and regulatory partners
Identifies conformational changes induced by phosphorylation
Provides structural insights complementary to antibody detection
Phosphoproteomics workflow integration:
Enrichment of phosphopeptides using TiO2 or immobilized metal affinity chromatography
High-resolution LC-MS/MS analysis
Parallel antibody validation with the same samples
Thermal Proteome Profiling (TPP):
Measures changes in protein thermal stability upon modification
Can detect functional consequences of PP2AA1 phosphorylation
Provides proteome-wide context for PP2AA1 regulation
These approaches provide antibody-independent confirmation of PP2AA1 phosphorylation status and offer deeper insights into the functional consequences of these modifications.
PP2AA1 antibodies are invaluable tools for studying complex PP2A assemblies:
PP2A holoenzyme composition analysis:
Immunoprecipitation with PP2AA1 antibodies followed by mass spectrometry
Identification of regulatory B subunits associated with specific complexes
Quantification of complex stoichiometry in different cellular contexts
STRIPAK complex investigation:
Dynamic complex assembly studies:
Time-course immunoprecipitation following stimulus
Tracking changes in complex composition during cellular responses
Correlation with functional outcomes
Spatial organization analysis:
Immunofluorescence with PP2AA1 antibodies reveals subcellular localization
Co-localization studies with other STRIPAK components
Super-resolution microscopy for nanoscale organization
Functional studies of assembled complexes:
Activity assays of immunopurified complexes
Reconstitution experiments with purified components
Structure-function analysis of mutant complexes
These approaches contribute to understanding how PP2AA1 functions within larger molecular assemblies to regulate diverse cellular processes including Hippo, MAPK, and cytoskeleton remodeling pathways .
Researchers should prioritize:
Rigorous validation:
Never rely solely on manufacturer claims about antibody specificity
Implement genetic controls (knockdown/knockout)
Use multiple antibodies targeting different epitopes
Be especially cautious with antibodies targeting post-translational modifications
Application-specific optimization:
Validate each antibody for specific applications (WB, IP, IF, IHC)
Optimize conditions including fixation, blocking, and antibody concentration
Consider that an antibody working well in one application may not work in others
Literature awareness:
Complementary approaches:
Supplement antibody-based detection with orthogonal methods
Consider mass spectrometry for definitive determination of post-translational modifications
Use functional assays to correlate antibody signals with biological activity
Detailed reporting:
Document all validation steps in publications
Report catalog numbers, clone information, and experimental conditions
Share validation data to benefit the research community
These considerations ensure robust, reproducible research with PP2AA1 antibodies and contribute to addressing the broader "reproducibility crisis" in biomedical research .
Innovative approaches hold promise for overcoming current challenges:
Recombinant antibody technology:
Generation of recombinant antibodies with precisely defined binding characteristics
Elimination of batch-to-batch variation through recombinant production
Engineering of antibodies with enhanced specificity for post-translational modifications
Single-domain antibodies (nanobodies):
Smaller size enables access to epitopes inaccessible to conventional antibodies
Potential for improved specificity for phosphorylated residues
Applications in live-cell imaging of dynamic PP2AA1 regulation
Proximity labeling approaches:
Antibody-enzyme fusions for proximity-dependent labeling
Mapping of PP2AA1 interaction networks in living cells
Temporal resolution of dynamic complex formation
Intrabodies and biosensors:
Expression of engineered antibody fragments in living cells
Real-time monitoring of PP2AA1 modifications and interactions
Correlation with functional outcomes in intact systems
Synthetic binding proteins:
Designed proteins with high specificity for particular PP2AA1 forms
Rational engineering based on structural information
Potential for absolute specificity for phosphorylated versus unphosphorylated forms