The Phospho-TP53 (Ser15) antibody recognizes a conserved epitope surrounding phosphorylated Ser15, with confirmed reactivity in human, mouse, rat, and monkey samples . Sequence homology predictions suggest potential cross-reactivity in pig, bovine, sheep, and rabbit tissues, though empirical validation remains pending . Importantly, the antibody does not bind non-phosphorylated p53, ensuring specificity in detecting the activated form of the protein . This specificity is critical for distinguishing basal p53 levels from stress-induced activation in experimental models .
Western blotting using this antibody typically employs dilutions ranging from 1:500 to 1:4000, depending on the cell lysate abundance and detection system sensitivity. For example:
These protocols consistently demonstrate increased Ser15 phosphorylation following genotoxic insults, correlating with p53 stabilization and nuclear accumulation .
For ChIP assays, 5 µL of antibody combined with 10 µg of chromatin (≈4×10⁶ cells) reliably enriches p53-bound DNA regions, such as promoters of target genes like p21 . The SimpleChIP® enzymatic kit validation confirms its utility in mapping p53’s transcriptional targets under DNA damage conditions .
In fixed HeLa cells, a 1:200–1:800 dilution localizes phospho-Ser15-p53 to the nucleus, with signal intensity proportional to UV dose . Co-staining with markers like tubulin (1:1000, NorthernLights™ secondary antibodies) enables spatial resolution of p53 activation relative to cytoskeletal changes .
Studies using this antibody have elucidated the temporal dynamics of Ser15 phosphorylation. In UV-treated HeLa cells, phospho-Ser15-p53 levels peak within 2–4 hours post-irradiation, preceding p21 induction and G1 arrest . Similarly, etoposide-treated HT-29 cells show sustained Ser15 phosphorylation, correlating with apoptosis via Bax upregulation . These findings underscore the antibody’s role in dissecting stress-specific p53 activation pathways.
In TP53-mutant cancers (e.g., Li-Fraumeni syndrome), the antibody detects residual Ser15 phosphorylation in truncated p53 isoforms, suggesting kinase activation independent of canonical DNA damage signaling . Such insights inform strategies to reactivate mutant p53 using MDM2 inhibitors or phosphorylation-stabilizing compounds .
Elevated Ser15 phosphorylation is observed in breast, colorectal, and glioblastoma tissues, often associated with chemotherapy resistance . A 2025 study using the HTRF® assay (Revvity) quantified phospho-Ser15-p53 in serum exosomes, proposing it as a non-invasive biomarker for tumor aggressiveness .
In neuronal models, oxidative stress-induced Ser15 phosphorylation correlates with synaptic plasticity deficits, implicating p53 in age-related cognitive decline . The antibody’s ability to detect low-abundance phospho-p53 in post-mitotic cells has advanced these investigations .
Essential controls include:
Lambda-phosphatase treatment: Pre-incubation with 600 U lambda-phosphatase for 1 hour should abolish the 53 kDa band .
Isoform specificity: Co-staining with pan-p53 antibodies (e.g., HAF1355) confirms phosphorylation-specific signals .
Cell line validation: Use etoposide/UV-treated HT-29 or HeLa cells as positive controls .
The HTRF® platform (Revvity) enables kinase inhibitor screening at physiological ATP concentrations, leveraging the antibody’s compatibility with biotinylated/6xHis-tagged substrates . This assay format has identified novel ATM/ATR inhibitors currently in preclinical trials .
Combining Ser15 detection with antibodies targeting Ser20, Ser37, or Ser392 could resolve spatial-temporal phosphorylation patterns, refining predictive models of p53-dependent outcomes .
Applications : Western blotting
Sample type: cells
Review: FF attenuation of CDDP cytotoxicity was secondary to Nrf2-dependent antioxidant generation but not to the modulation of p53-dependent DNA damage response.
Phosphorylation of p53 at Serine 15 (Ser15) is a critical post-translational modification that regulates p53 activity in response to cellular stress. This phosphorylation event significantly influences p53's tumor suppressor functions through multiple mechanisms:
It disrupts the interaction between p53 and its negative regulator MDM2, preventing ubiquitination and proteasomal degradation of p53
It enhances p53-dependent transcription from responsive promoters including p21, BAX, and MDM2
It is required for the recruitment of p53 to promoters post-stimulus
It promotes local histone acetylation and chromatin relaxation, facilitating transcriptional activation
Studies have demonstrated that this phosphorylation occurs both after DNA damage and at basal levels in unstimulated cells, indicating its importance in both stress-induced and physiological p53 function .
Several methodologies are available for detecting Phospho-p53 (Ser15), each with distinct advantages depending on your research questions:
For experiments requiring high sensitivity, HTRF and MSD-based electrochemiluminescence assays provide excellent signal-to-noise ratios for detecting subtle changes in phosphorylation levels .
Validating antibody specificity is crucial for reliable experimental outcomes. A comprehensive validation approach should include:
Positive and negative controls: Use UV-irradiated cells (40 mJ/cm²) as positive controls, which show significant increase in p53-Ser15 phosphorylation within 1 hour post-treatment . Untreated cells serve as negative controls.
Phosphorylation-site mutants: Test antibody reactivity against p53 S15A (alanine substitution) and S15D (phospho-mimic) mutants. A specific antibody will not recognize S15A but may recognize S15D .
Phosphatase treatment: Treat positive control samples with lambda phosphatase; this should eliminate signal from a phospho-specific antibody.
Western blot analysis: Verify a single band at the expected molecular weight (53 kDa) and compare with total p53 antibody blotting .
Immunofluorescence:
For comprehensive validation, compare results across multiple detection methods to confirm consistent patterns of phosphorylation in response to stimuli .
Optimizing ChIP protocols for Phospho-p53 (Ser15) requires careful consideration of multiple parameters:
Antibody concentration: For optimal ChIP results, use 5 μL of antibody and 10 μg of chromatin (approximately 4 × 10^6 cells) per immunoprecipitation . This ratio ensures sufficient antibody for complete capture without excess that could increase background.
Crosslinking conditions: Since phosphorylated p53 may have different DNA binding dynamics than total p53, optimize formaldehyde crosslinking time (typically 10-15 minutes at room temperature) to capture transient interactions.
Sonication parameters: Phospho-p53 (Ser15) has been shown to influence local chromatin structure , so optimize sonication conditions to achieve chromatin fragments of 200-500 bp while preserving epitope integrity.
Washing stringency: Balance between removing non-specific binding while maintaining specific interactions; phospho-epitopes may be more sensitive to high-salt conditions.
Elution and reversal: Use enzymatic chromatin IP kits which have been validated with this antibody for optimal results.
Controls: Include:
After ChIP, qPCR primer design should target regions containing p53 response elements in promoters of interest, with special attention to p21/CDKN1A which shows strong dependency on Ser15 phosphorylation .
Accurate quantification of p53-Ser15 phosphorylation relative to total p53 requires careful methodological consideration:
Signal normalization strategy: Calculate the ratio of phosphorylated p53 signal increase (Δp53-Ser15P) to total p53 increase (Δp53) . This approach distinguishes between:
Simple accumulation of total p53 (where the phosphorylation proportion remains unchanged)
Actual increases in the phosphorylation stoichiometry
Cell cycle considerations: Phosphorylation levels can vary significantly by cell cycle phase:
Time course analysis: Maximum p53-Ser15 phosphorylation typically occurs 4-6 hours after DNA damage stimulus, which coincides with peak Chk2 activation but not ATM activation and H2AX phosphorylation (which typically peak 1-2 hours post-treatment) .
Antibody selection: Use antibody pairs validated to work together in multiplex assays that simultaneously detect phosphorylated and total p53 .
Sample preparation: Standardize lysis conditions to ensure complete extraction of both nuclear (where most phosphorylated p53 resides) and cytoplasmic p53 pools.
A well-designed experiment should include western blot validation alongside quantitative assays to confirm that changes in signaling are not artifacts of detection method .
Distinguishing between ATM-dependent and Chk2-dependent phosphorylation of p53 at Ser15 requires a multi-faceted experimental approach:
Kinetic analysis: Based on temporal patterns, design time-course experiments:
Specific inhibitors:
KU-55933 (ATM inhibitor): Blocks early phosphorylation events
Chk2 Inhibitor II: Targets later phosphorylation events
Compare the inhibition patterns to determine the relative contribution of each kinase
Genetic approaches:
siRNA/shRNA knockdown of ATM or Chk2
CRISPR/Cas9-mediated knockout cells
Complementation with kinase-dead mutants
Cellular localization studies:
Stimulus-specific analysis:
A comprehensive phospho-proteomic approach using mass spectrometry can further distinguish the exact contribution of each kinase by identifying co-occurring phosphorylation events specific to each pathway .
A robust experimental design for measuring p53-Ser15 phosphorylation should include these essential controls:
Positive controls:
UV-irradiated cells (40 mJ/cm²): These exhibit strong p53-Ser15 phosphorylation within 1 hour of treatment
Topoisomerase inhibitor-treated cells: TPT or MXT treatment for 4-6 hours induces significant phosphorylation
Etoposide treatment: Creates double-strand breaks that trigger ATM-dependent phosphorylation
Negative controls:
Specialized controls:
Nutlin-3a treated cells: Causes p53 accumulation with minimal increase in Ser15 phosphorylation stoichiometry
IPTG-inducible p53 expression systems: Allow comparison of wild-type p53 with S15A and S15D mutants at identical expression levels
Cell cycle synchronized populations: Important because mitotic cells show constitutively high p53-Ser15 phosphorylation
Technical controls:
When interpreting results, remember that basal phosphorylation levels are normally present in unstimulated cells and play physiological roles, rather than representing background or non-specific signal .
Resolving contradictory results between different detection methods requires systematic analysis of methodological differences:
Epitope accessibility differences:
Western blotting: Denatured proteins expose all epitopes
Immunoprecipitation/ChIP: Conformational changes or protein-protein interactions may mask epitopes
ELISA/HTRF: Sandwich antibody configurations require simultaneous binding of two antibodies
Solution: Use different antibody clones targeting the same phospho-epitope but with different binding characteristics.
Sample preparation effects:
Phosphatase activity during lysis can reduce signal
Different lysis buffers extract different subcellular pools of p53
Nuclear extraction efficiency varies between methods
Solution: Include phosphatase inhibitors in all buffers; standardize extraction protocols; compare nuclear and cytoplasmic fractions separately.
Signal calibration issues:
Western blot: Semi-quantitative; depends on exposure time
ELISA/HTRF: Quantitative but may have different dynamic ranges
Solution: Generate standard curves using recombinant phosphorylated and non-phosphorylated p53; include gradient of positive control samples.
Cell heterogeneity effects:
Population-based assays (Western blot, ELISA): Average signal across all cells
Single-cell methods (Flow cytometry, immunofluorescence): Reveal subpopulations
Solution: Compare cell cycle-synchronized populations; use flow cytometry to isolate subpopulations for further analysis .
Temporally dynamic phosphorylation:
Different methods may have different processing times
Solution: Standardize sample handling time; perform time-course experiments.
When methods consistently disagree, consider that each may be measuring different pools of phosphorylated p53 with different biological significance .
Working with Phospho-p53 (Ser15) antibodies in fixed tissue samples presents several technical challenges that require specific optimization strategies:
Epitope masking during fixation:
Formalin fixation can cross-link proteins and mask phospho-epitopes
Solution: Optimize antigen retrieval methods; compare heat-induced (citrate buffer, pH 6.0) versus enzyme-based retrieval; test different retrieval times (10-30 minutes).
Phosphatase activity during tissue processing:
Delay between tissue collection and fixation can reduce phospho-signals
Solution: Ensure rapid fixation of tissues; include phosphatase inhibitors in buffers; consider using PAXgene or other phospho-preserving fixatives.
Non-specific background in immunohistochemistry:
Endogenous peroxidase activity can cause false positives
Solution: Include proper blocking steps (hydrogen peroxide block, protein block); optimize antibody concentration (typically start at 10-15 μg/mL) ; include appropriate controls.
Tissue heterogeneity and interpretation challenges:
Variable fixation across tissue sections
Mixed cell populations within samples
Solution: Use multi-staining approaches to identify specific cell types; quantify signal intensity using digital image analysis.
Quantification limitations:
Chromogenic IHC has limited dynamic range for quantification
Solution: Consider fluorescent IHC for better quantification; always include a range of control samples with known phosphorylation status in each batch.
Validation across species:
Antibody may have different performance across species
Solution: Verify species cross-reactivity; the p53-Ser15 epitope is highly conserved across human, mouse and rat, but testing is advised .
When analyzing human cancer tissue samples, remember that p53 mutations may affect antibody binding or phosphorylation patterns, so correlation with molecular data on p53 mutation status can help interpretation .
The differential impact of p53-Ser15 phosphorylation on cellular fate decisions involves complex regulatory mechanisms:
Promoter-specific effects:
Cell cycle arrest genes (p21/CDKN1A): Studies show complete dependence on Ser15 phosphorylation for activation
Apoptotic genes (BAX): Show reduced but not abolished activation with S15A mutation
This differential requirement suggests Ser15 phosphorylation may preferentially direct p53 toward cell cycle arrest programs.
Threshold effects:
Temporal dynamics:
Early/transient Ser15 phosphorylation tends to promote cell cycle arrest
Sustained phosphorylation, especially when combined with other modifications, shifts the balance toward apoptosis
Cooperation with other modifications:
Cell type-specific responses:
Experimental evidence suggests that S15D phospho-mimic mutations can rescue transcriptional activity at all p53-responsive promoters, indicating that the phosphorylation is necessary but works in concert with other modifications to determine final cellular outcomes .
The relationship between p53-Ser15 phosphorylation and histone modifications represents a critical mechanistic link in transcriptional regulation:
Recruitment of histone acetyltransferases (HATs):
Chromatin relaxation mechanism:
ChIP analyses reveal that S15A mutant p53 binds to promoters but fails to initiate local histone acetylation
Ser15 phosphorylation is required for chromatin relaxation at p53-responsive promoters
This chromatin remodeling is necessary for recruitment of RNA polymerase II and transcriptional machinery
Sequential modification model:
Ser15 phosphorylation occurs first
This enables p53 to recruit HATs
HATs acetylate both p53 itself (enhancing DNA binding) and local histones
The resulting open chromatin structure allows additional transcription factors to bind
Promoter-specific effects:
Cell cycle-dependent patterns:
To experimentally investigate this relationship, ChIP-reChIP experiments (sequential immunoprecipitation) can be performed using anti-phospho-p53(Ser15) antibody followed by antibodies against modified histones (e.g., H3K9ac, H3K4me3) to determine their co-occurrence at specific genomic loci .
Different DNA-damaging agents produce distinct patterns of p53-Ser15 phosphorylation through varied mechanisms:
Topoisomerase inhibitors:
Radiation-induced damage:
Chemical agents:
Alkylating agents: Slower phosphorylation kinetics
Crosslinking agents: Variable patterns depending on repair pathway activation
Replication inhibitors: S-phase specific phosphorylation
Non-genotoxic p53 activators:
Kinase pathway specificity:
ATM-dependent phosphorylation: Predominant after double-strand breaks
ATR-dependent phosphorylation: Major pathway after UV damage and replication stress
DNA-PK-dependent phosphorylation: Contributes following certain types of DNA damage
Chk1/Chk2-dependent phosphorylation: Secondary wave following ATM/ATR activation
For experimental design, consider that the ratio of phosphorylated to total p53 (Δp53-Ser15P/Δp53) provides more meaningful data than absolute phosphorylation levels, as it distinguishes between simple p53 accumulation and increased phosphorylation stoichiometry .
Integrating Phospho-p53 (Ser15) detection into multiplexed phospho-proteomic workflows requires strategic experimental design:
Mass spectrometry-based approaches:
Enrichment strategies: Use antibodies against Phospho-p53 (Ser15) for immunoprecipitation prior to MS analysis
Targeted MS methods: Develop Multiple Reaction Monitoring (MRM) or Parallel Reaction Monitoring (PRM) assays specific for p53-Ser15 phosphopeptides
Internal standards: Include synthetic phosphopeptides containing the Ser15 site as quantitative references
Multiplexed antibody-based detection:
HTRF technology: Allows simultaneous detection of phospho-p53 and total p53
MSD platform: Electrochemiluminescence-based detection enables multiplexing of phospho-p53 with other phospho-proteins in the same pathway
Multiplex flow cytometry: Combine Phospho-p53 (Ser15) antibodies with markers for cell cycle, DNA damage (γH2AX), and apoptosis
High-content imaging approaches:
Multiplex immunofluorescence: Combine with other phospho-epitopes (ATM, Chk2, H2AX)
Quantitative image analysis: Measure nuclear:cytoplasmic ratios, foci formation, and co-localization with DNA damage markers
Single-cell applications:
Mass cytometry (CyTOF): Incorporate metal-tagged Phospho-p53 (Ser15) antibodies into panels with other phospho-proteins
Single-cell phospho-proteomics: Emerging techniques for measuring phosphorylation in individual cells
Temporal dynamics analysis:
Live-cell reporters: Develop FRET-based sensors to monitor p53-Ser15 phosphorylation in real-time
Kinetic measurements: Establish time-resolved assays to capture phosphorylation dynamics
When designing multiplexed assays, ensure antibody compatibility (species, isotypes) and validate that detection antibodies do not compete for overlapping epitopes. Cross-validate findings with orthogonal methods to confirm specificity in complex samples .
Phospho-p53 (Ser15) antibodies are finding increasing utility in clinical applications:
Predictive biomarker applications:
Treatment response prediction: p53-Ser15 phosphorylation levels can predict sensitivity to DNA-damaging chemotherapeutics
Resistance mechanisms: Defective phosphorylation pathways correlate with treatment resistance
Patient stratification: Different patterns of phosphorylation may identify patients likely to benefit from specific therapies
Pharmacodynamic monitoring:
Early response assessment: Measure p53-Ser15 phosphorylation as an early pharmacodynamic marker of drug activity
Optimal timing: Establish time-dependent patterns after treatment to determine optimal assessment windows
Minimal residual disease: Monitor restoration of normal p53 signaling after therapy
Liquid biopsy developments:
Circulating tumor cells (CTCs): Analysis of p53-Ser15 phosphorylation in CTCs as a minimally invasive biomarker
Extracellular vesicles: Detection of phospho-p53 in tumor-derived exosomes
Cell-free DNA studies: Correlation between circulating tumor DNA and phospho-p53 status in tumor tissues
Advanced tissue diagnostics:
Multiplex immunohistochemistry: Combined detection of phospho-p53 with other markers
Digital pathology: Automated quantification of nuclear phospho-p53 staining
Spatial transcriptomics integration: Correlate phospho-p53 status with local gene expression patterns
Therapeutic targeting applications:
Monitoring ATM/ATR inhibitor efficacy: Changes in p53-Ser15 phosphorylation as target engagement biomarker
Combination therapy rationale: Identify synergistic combinations based on phosphorylation patterns
Synthetic lethality approaches: Target cells with specific p53 phosphorylation defects
Recent studies have shown that analyzing p53-Ser15 phosphorylation patterns in breast cancer tissue can provide prognostic information beyond p53 mutation status alone , suggesting value in incorporating this marker into routine cancer diagnostics.
Designing experiments to elucidate the interplay between p53-Ser15 phosphorylation and other post-translational modifications requires sophisticated methodological approaches:
Sequential modification studies:
Time-course experiments: Track the order of appearance of different modifications
Site-directed mutagenesis: Create Ser15 phospho-mimetic (S15D) or phospho-dead (S15A) mutants and analyze effects on other modifications
Inducible expression systems: Control p53 expression level to normalize comparisons between wild-type and mutant proteins
Modification-specific antibody combinations:
Multiplex immunoassays: Simultaneously detect multiple modifications (phosphorylation, acetylation, methylation, ubiquitination)
Sequential immunoprecipitation (IP-reIP): Use phospho-p53 (Ser15) antibody for first IP, followed by antibodies against other modifications
Proximity ligation assays: Detect co-occurrence of multiple modifications on the same p53 molecule
Mass spectrometry-based approaches:
Top-down proteomics: Analyze intact p53 to preserve modification combinations
Middle-down strategies: Analyze large p53 fragments to maintain modification patterns
Targeted MS: Develop assays for specific combinations of modifications
Crosslinking MS: Identify interaction partners specific to phosphorylated p53
Functional correlation experiments:
Mathematical modeling approaches:
Develop kinetic models of p53 modification networks
Predict and test modification dependencies and hierarchies
Integrate experimental data with computational approaches to understand emergent properties
To specifically study interactions between Ser15 phosphorylation and other key modifications, consider: