PP2A’s B subunit exists in multiple isoforms (e.g., B55α, B56γ), which dictate the enzyme’s substrate selectivity and cellular functions. The B subunit facilitates interactions with substrates like AKT, β-catenin, and components of the mTOR pathway, influencing processes such as cell cycle progression, apoptosis, and immune responses . Dysregulation of PP2A-B subunit interactions is linked to cancers, neurodegenerative diseases, and autoimmune disorders .
The PP2A B Subunit (100C1) Rabbit mAb (#2290) exhibits the following properties :
Property | Detail |
---|---|
Host Species | Rabbit |
Clonality | Monoclonal (Clone 100C1) |
Applications | Western Blot (WB), Immunoprecipitation (IP), Immunohistochemistry (IHC) |
Species Reactivity | Human, Mouse, Rat, Monkey, D. melanogaster |
Molecular Weight | 52 kDa |
Catalog Number | #2290 (Cell Signaling Technology) |
This antibody detects endogenous PP2A B subunits without cross-reacting with other phosphatase regulatory subunits .
PP2A-B subunit antibodies have been instrumental in identifying PP2A’s tumor-suppressive roles. For example:
PP2A-B subunit dysfunction promotes hyperactivation of oncogenic pathways like PI3K/AKT/mTOR and WNT/β-catenin, accelerating tumor progression .
In lung cancer models, PP2A-B subunit mutants (e.g., Aα-E64D) correlate with reduced survival and enhanced RAS-driven transformation .
PP2A-deficient B cells show impaired germinal center formation and reduced responses to T cell-dependent antigens, highlighting its role in adaptive immunity .
Transcriptomic analyses reveal that PP2A regulates NAD metabolism and purine/pyrimidine pathways in B cells, linking its activity to metabolic reprogramming in autoimmunity .
PP2A (Protein Phosphatase 2A) is a major serine/threonine phosphatase involved in regulating diverse cellular processes including signal transduction, cell cycle progression, and metabolism. The PP2A holoenzyme consists of a catalytic subunit (C), a scaffold subunit (A), and a regulatory subunit (B). PP2AB1 represents a specific regulatory B subunit that directs the phosphatase activity to particular substrates and cellular locations. This specificity is crucial for cellular function as PP2A is involved in the regulation of a wide variety of enzymes, signal transduction pathways, and cellular events . The B subunits are particularly important as they determine substrate specificity and subcellular localization of the PP2A holoenzyme complex.
PP2A antibodies vary significantly in their epitope recognition and specificity for different PP2A subunits. Anti-PP2A antibodies typically detect proteins encoded by genes such as PTPA (protein phosphatase 2 phosphatase activator) . Some antibodies recognize the catalytic subunits (PP2A-alpha and PP2A-beta), while others detect specific regulatory subunits like PP2AB1. Antibodies may also be designed to recognize post-translationally modified forms, such as phosphorylated PP2A at Y307 . When selecting an antibody, researchers should carefully evaluate whether they need an antibody that recognizes multiple PP2A isoforms or one with high specificity for PP2AB1 only, depending on their experimental goals.
PP2AB1 antibodies are employed across multiple experimental techniques including:
Western blot (WB): For detecting and quantifying PP2AB1 protein levels in cell and tissue lysates
Immunohistochemistry (IHC): For visualizing PP2AB1 distribution in tissue sections
Immunocytochemistry/Immunofluorescence (ICC/IF): For determining subcellular localization
Flow cytometry (FCM): For quantifying PP2AB1 in individual cells
Immunoprecipitation (IP): For isolating PP2A complexes containing PP2AB1
These applications are particularly valuable for studying PP2A's roles in cytoskeleton remodeling, cell signaling pathways, and its interactions with other proteins in various cellular contexts.
Validating antibody specificity is critical for reliable results. A comprehensive validation approach should include:
Positive and negative controls: Use tissues or cell lines known to express high levels of PP2AB1 (positive control) and those with minimal or no expression (negative control).
Knockout/knockdown validation: Compare antibody reactivity in wild-type samples versus those where PP2AB1 has been knocked out or knocked down using siRNA/shRNA.
Peptide competition assay: Pre-incubate the antibody with the immunizing peptide before application to samples. If specific, the signal should be significantly reduced or eliminated.
Cross-reactivity testing: Test the antibody against other PP2A subunits, particularly other B-family members, to ensure specificity to PP2AB1.
Multiple antibody comparison: Use multiple antibodies targeting different epitopes of PP2AB1 and compare the results for consistency .
When designing experiments with PP2AB1 antibodies, include these critical controls:
Loading controls: Use housekeeping proteins (β-actin, GAPDH) for Western blots to normalize protein loading.
Isotype controls: Include appropriate isotype-matched negative control antibodies to assess non-specific binding.
Secondary antibody only: Include samples treated with only secondary antibody to evaluate background.
Positive biological controls: Include samples known to express PP2AB1 at different levels to establish signal linearity.
Phosphorylation state controls: When studying phosphorylation-dependent interactions, include phosphatase-treated and untreated samples to verify phosphorylation-specific effects .
Recombinant protein standards: Include purified recombinant PP2AB1 as a standard for size verification and quantification.
To effectively detect PP2AB1-containing complexes:
Lysis buffer optimization: Use buffers that maintain protein-protein interactions. Avoid harsh detergents that may disrupt PP2A holoenzyme integrity. Phosphatase inhibitors are crucial to preserve the phosphorylation state of PP2A and its targets.
Cross-linking approaches: Consider mild cross-linking before lysis to stabilize transient interactions.
Immunoprecipitation conditions: Optimize antibody concentration, incubation time and temperature, and washing stringency to maintain specific interactions while reducing background.
Native gel electrophoresis: Consider native PAGE instead of denaturing SDS-PAGE when studying intact complexes.
Size exclusion chromatography: Use this as a complementary approach to isolate PP2A complexes based on molecular weight before immunological detection .
STRIPAK complex analysis: When studying PP2A in the context of striatin-interacting phosphatase and kinase (STRIPAK) complexes, consider specialized approaches as these complexes regulate multiple signaling pathways including Hippo, MAPK, and cytoskeleton remodeling pathways .
For optimal Western blot results with PP2AB1 antibodies:
Sample preparation:
Extract proteins using RIPA or NP-40 buffer supplemented with protease and phosphatase inhibitors
Include 1-5 mM sodium fluoride and 1 mM sodium orthovanadate to preserve phosphorylation states
Heat samples at 95°C for 5 minutes in Laemmli buffer with reducing agent
Gel electrophoresis and transfer:
Use 10-12% polyacrylamide gels for optimal resolution of PP2AB1 (expected size varies by species)
Transfer to PVDF membranes at 100V for 60-90 minutes in cold transfer buffer containing 10-20% methanol
Antibody incubation:
Block membranes in 5% non-fat milk or BSA (particularly for phospho-specific antibodies) for 1 hour at room temperature
Dilute primary antibody at 1:1000 (typical range 1:500-1:2000, optimize for each antibody)
Incubate overnight at 4°C with gentle rocking
Wash 3-5 times with TBST
Incubate with appropriate HRP-conjugated secondary antibody at 1:5000 for 1 hour at room temperature
Wash thoroughly before detection
Detection and analysis:
For successful immunofluorescence with PP2AB1 antibodies:
Fixation methods:
Permeabilization:
Use 0.1-0.2% Triton X-100 in PBS for 5-10 minutes
For phospho-epitopes, consider 0.5% saponin which is sometimes less disruptive
Blocking and antibody dilutions:
Co-staining strategies:
Imaging considerations:
Use confocal microscopy for precise subcellular localization
Employ deconvolution for improved resolution
Consider super-resolution techniques for detailed study of PP2A complex localization
When encountering problems with PP2AB1 antibodies, consider these troubleshooting strategies:
Issue | Possible Causes | Troubleshooting Approaches |
---|---|---|
No signal | Antibody degradation, low protein expression, inappropriate application | Test antibody with positive control samples, optimize protein extraction, try alternative detection methods |
Multiple bands | Cross-reactivity, protein degradation, post-translational modifications | Use more specific antibody, add protease inhibitors, verify with knockout samples |
High background | Insufficient blocking, antibody concentration too high, inadequate washing | Increase blocking time/concentration, titrate antibody, increase wash duration/stringency |
Inconsistent results | Lot-to-lot variation, sample heterogeneity, protocol inconsistency | Use same antibody lot when possible, standardize protocols, include internal controls |
Weak signal | Low protein abundance, inefficient extraction, epitope masking | Increase sample concentration, optimize extraction method, try alternative epitope antibodies |
Additionally, phosphorylation of PP2A at Y307 can affect antibody recognition and enzymatic activity . If investigating phosphorylated forms, consider phosphatase treatments of control samples to verify signal specificity.
Post-translational modifications (PTMs) significantly influence PP2AB1 antibody binding efficacy:
Phosphorylation effects:
Phosphorylation at Y307 of the catalytic subunit is a key regulatory modification that can mask epitopes recognized by certain antibodies
This phosphorylation inhibits PP2A activity, making phospho-specific antibodies valuable for studying PP2A regulation
When studying PP2A activity, consider using both phospho-specific and total protein antibodies
Methylation considerations:
C-terminal leucine methylation of PP2A catalytic subunit affects regulatory subunit binding
If studying holoenzyme assembly, verify whether your antibody's recognition is affected by methylation status
Other modifications:
Ubiquitination can target PP2A for degradation and may interfere with antibody binding
Oxidative modifications can occur during sample processing and alter epitope recognition
Detecting modified forms:
Recent advances in studying PP2A-mediated signaling include:
Proximity labeling approaches:
BioID or APEX2 fusion proteins with PP2AB1 can identify proximal interaction partners
These methods capture both stable and transient interactions in living cells
Combine with PP2AB1 antibodies for verification of identified interactions
Live-cell imaging techniques:
FRET-based sensors can monitor PP2A activity in real-time
PP2AB1 antibody fragments (Fabs) conjugated to fluorophores for live tracking
Verification of FRET results with fixed-cell antibody staining provides complementary data
ChIP-derived techniques:
For studying PP2A's role in transcriptional regulation
PP2AB1 antibodies can be used in ChIP-seq experiments to identify genomic regions where PP2A complexes associate
Correlate with transcriptional changes when PP2A activity is modulated
Interaction network mapping:
Combine immunoprecipitation with mass spectrometry (IP-MS)
Use PP2AB1 antibodies to pull down PP2A complexes and identify associated proteins
Compare interaction networks under different cellular conditions or treatments
STRIPAK complex analysis:
To investigate specific PP2A-substrate relationships:
Substrate-trapping approaches:
Use catalytically inactive PP2A mutants to trap substrates
Immunoprecipitate with PP2AB1 antibodies and identify bound substrates
Confirm interactions with reciprocal co-immunoprecipitation experiments
Phosphoproteomics integration:
Compare phosphoproteomic profiles before and after PP2A inhibition/activation
Use bioinformatics to identify putative substrates with PP2A consensus motifs
Validate candidates using PP2AB1 antibodies in targeted experiments
In vitro dephosphorylation assays:
Immunopurify PP2A holoenzymes containing PP2AB1 using specific antibodies
Test dephosphorylation activity on candidate substrates
Measure dephosphorylation kinetics under various conditions
Localization-based studies:
Specific pathway investigation:
PP2A regulates multiple specific pathways, including:
When facing contradictory results with different PP2AB1 antibodies:
Epitope mapping analysis:
Determine the specific epitopes recognized by each antibody
Epitopes in regions affected by post-translational modifications or protein-protein interactions may yield differential results
Antibodies recognizing different domains may detect different subpopulations of PP2AB1
Validation hierarchy assessment:
Prioritize results from antibodies validated through multiple approaches (knockout controls, recombinant protein testing)
Consider the validation status of each antibody and its history in the literature
Antibodies with published validation in your specific application deserve higher confidence
Technical vs. biological variability:
Resolution strategies:
Use complementary non-antibody approaches (mass spectrometry, CRISPR/Cas9 tagging)
Combine multiple antibodies recognizing different epitopes in the same experiment
When reporting contradictory results, include data from all antibodies tested with transparent discussion of limitations
For accurate quantification of PP2AB1 expression:
Standard curve calibration:
Use recombinant PP2AB1 protein standards to create a calibration curve
Ensure the standard curve covers the expected range of expression in samples
Include standards in each experimental run to account for inter-assay variability
Normalization approaches:
For Western blots, normalize to appropriate loading controls (β-actin, GAPDH)
For tissue samples, consider cell-type specific normalization if PP2AB1 expression varies by cell type
In phosphorylation studies, present data as the ratio of phosphorylated to total protein
Quantification methods:
For fluorescence-based detection, ensure measurements are in the linear range
For chemiluminescence, use multiple exposure times to prevent signal saturation
Use digital image analysis software with background subtraction
Statistical analysis requirements:
Run sufficient biological replicates (minimum n=3, preferably n≥5)
Apply appropriate statistical tests based on data distribution
Report variability (standard deviation or standard error) along with means
Technical considerations:
Account for antibody affinity differences when comparing different PP2A subunits
Be aware that total PP2A levels may not reflect active enzymes due to post-translational modifications
Consider the impact of sample preparation on epitope preservation
The reliability of PP2AB1 antibodies for interaction studies varies by context:
Co-immunoprecipitation (Co-IP) reliability:
Generally high reliability for stable interactions within the PP2A holoenzyme
Moderate reliability for dynamic or transient interactions
Effectiveness depends on antibody quality, epitope accessibility, and buffer conditions
Critical to verify that the antibody does not compete with interacting proteins for binding
Proximity ligation assay (PLA) considerations:
Higher sensitivity than co-IP for detecting in situ interactions
Requires validation of both PP2AB1 antibody and partner protein antibody
False positives can occur if proteins are in close proximity but not directly interacting
Useful for confirming interactions suggested by co-IP or yeast two-hybrid screens
Experimental system variations:
Cell line models: Generally provide consistent results but may not recapitulate tissue-specific interactions
Tissue samples: Offer physiological relevance but present challenges due to cellular heterogeneity
In vitro systems with purified components: Highest specificity but may miss interactions dependent on cellular context
Validation strategies:
Reciprocal Co-IP (using antibodies against both interaction partners)
Competition with excess peptide corresponding to the antibody epitope
Demonstration that interaction is lost when binding sites are mutated
Correlation of interaction with functional outcomes
Specific PP2A interaction contexts:
Recent technological advances enhancing PP2AB1 antibody applications include:
Single-cell analysis techniques:
Single-cell Western blotting allows PP2AB1 quantification at individual cell level
Mass cytometry (CyTOF) with metal-conjugated antibodies enables high-dimensional analysis
These approaches reveal heterogeneity in PP2A expression and regulation previously masked in bulk analyses
Advanced imaging methodologies:
Super-resolution microscopy overcomes diffraction limits to visualize PP2A complexes at nanoscale
Expansion microscopy physically enlarges specimens for improved resolution with standard microscopes
Light-sheet microscopy enables rapid 3D imaging of PP2A distribution in intact tissues
Spatially-resolved proteomics:
Digital spatial profiling combines antibody detection with spatial coordinates
Allows mapping of PP2A complexes within tissue architecture
Integration with single-cell transcriptomics creates multi-omic spatial profiles
Engineered antibody formats:
Single-domain antibodies (nanobodies) against PP2AB1 for improved penetration and reduced interference
Bispecific antibodies targeting PP2AB1 and interacting partners simultaneously
Intrabodies for tracking PP2A in living cells without fixation artifacts
Computational integration:
Machine learning algorithms to detect subtle patterns in PP2A localization and interactions
Network analysis tools to place PP2A-centered interactions in broader signaling contexts
These computational approaches help interpret complex datasets generated using PP2AB1 antibodies
Researchers should consider these important limitations:
Cross-reactivity challenges:
High sequence homology between PP2A family members can limit absolute specificity
Some antibodies may not distinguish between highly similar isoforms
Cross-reactivity needs to be systematically evaluated in each experimental system
Conformational sensitivity:
Antibodies may preferentially recognize certain conformational states of PP2A
Incorporation into holoenzyme complexes may mask or expose different epitopes
Sample preparation can alter conformation and affect antibody recognition
Post-translational modification interference:
Technical variability:
Lot-to-lot variation remains a significant challenge with polyclonal antibodies
Storage conditions and freeze-thaw cycles can impact antibody performance
Protocol optimization often required when switching between antibody batches
Application-specific limitations:
Fixation-sensitive epitopes may be lost in certain applications (e.g., IHC)
Detergents required for extraction may disrupt important interactions
Species cross-reactivity varies, limiting use in certain model organisms
Future directions in PP2AB1 antibody research may include:
Development of conformation-specific antibodies:
Antibodies that specifically recognize active vs. inactive PP2A conformations
Tools to distinguish free catalytic subunits from those in holoenzyme complexes
Antibodies sensitive to allosteric changes induced by regulatory proteins
Integration with CRISPR technologies:
CRISPR knock-in of epitope tags to facilitate detection without relying on antibody specificity
Complementary approaches using endogenously tagged PP2A subunits alongside antibody detection
Validation systems using CRISPR-generated knockout controls
Enhancing temporal resolution:
Development of biosensors based on PP2AB1 antibody fragments
Systems to track PP2A localization and activity in real-time
Tools for acute inactivation or activation of specific PP2A complexes
Therapeutic applications:
Multi-omics integration:
Combining antibody-based detection with transcriptomics, metabolomics, and other -omics approaches
Systems biology frameworks to place PP2A signaling in broader cellular contexts
Machine learning integration to predict PP2A substrates and pathway impacts