The antibody is produced through a multi-step process:
Gene Extraction: Antibody-coding genes are extracted from rabbits immunized with a synthetic peptide corresponding to phosphorylated S33 on human TP53 .
Vector Transfection: Genes are cloned into expression vectors and transfected into suspension-adapted host cells (e.g., HEK293F) .
Expression and Purification: Recombinant antibodies are secreted into cell culture supernatant and purified via affinity chromatography .
Validation: Specificity is confirmed through ELISA, immunohistochemistry (IHC), and immunofluorescence (IF) .
Phosphorylation at S33 stabilizes p53, enabling its interaction with DNA repair machinery and transcriptional targets .
Validated in stress models: Etoposide-treated T-47D cells show increased nuclear staining, reversible by alkaline phosphatase treatment .
Cross-reactivity confirmed in human cell lines (e.g., A431, HeLa) and non-human primates .
Observed Band: 53 kDa (vs. predicted 43 kDa), consistent with post-translational modifications .
Lane-Specific Activity:
Nuclear localization of phosphorylated p53 increases in etoposide-treated A431 cells .
Co-staining with α-tubulin (Alexa Fluor® 594) validates subcellular specificity .
Cancer Studies: Detects aberrant p53-S33 phosphorylation in tumors, aiding in prognostic assessments .
Drug Development: Screens for compounds modulating p53 activity via S33 phosphorylation .
Mechanistic Insights: Links phosphorylation status to p53’s transcriptional activity in DNA damage response .
The Phospho-TP53 (S33) Recombinant Monoclonal Antibody is produced through a rigorous process that begins with the extraction of genes coding for the TP53 antibody from rabbits previously immunized with a synthetic peptide derived from the human TP53 protein phosphorylated at S33. These antibody genes are then incorporated into specialized expression vectors and meticulously transfected into host suspension cells. Following successful transfection, the positive cells are cultivated to facilitate the robust expression and secretion of the antibodies. The Phospho-TP53 (S33) Recombinant Monoclonal Antibody is subsequently purified using affinity chromatography techniques, isolating the antibody from the cell culture supernatant. The antibody's efficacy is rigorously assessed through a series of tests, including ELISA, IHC, and IF assays, confirming its ability to interact effectively with human TP53 protein phosphorylated at S33.
Phosphorylation of p53 at S33 plays a critical role in coordinating DNA repair, cell cycle regulation, and cell fate decisions in response to stress and damage. Disruptions in this phosphorylation event can lead to uncontrolled cell proliferation, a common occurrence in cancer cells.
TP53 acts as a tumor suppressor in various tumor types, inducing growth arrest or apoptosis depending on the specific physiological circumstances and cell type. It plays a crucial role in cell cycle regulation as a trans-activator that negatively regulates cell division by controlling genes essential for this process. One of the genes activated by TP53 is an inhibitor of cyclin-dependent kinases. Apoptosis induction appears to be mediated either through stimulation of BAX and FAS antigen expression, or by repressing Bcl-2 expression. Its pro-apoptotic activity is activated via its interaction with PPP1R13B/ASPP1 or TP53BP2/ASPP2. However, this activity is inhibited when the interaction with PPP1R13B/ASPP1 or TP53BP2/ASPP2 is displaced by PPP1R13L/iASPP. In collaboration with mitochondrial PPIF, TP53 is involved in activating oxidative stress-induced necrosis; this function is largely independent of transcription. TP53 induces the transcription of long intergenic non-coding RNA p21 (lincRNA-p21) and lincRNA-Mkln1. LincRNA-p21 participates in TP53-dependent transcriptional repression leading to apoptosis and appears to have an effect on cell-cycle regulation. TP53 is implicated in Notch signaling cross-over. It prevents CDK7 kinase activity when associated with the CAK complex in response to DNA damage, thus halting cell cycle progression. Isoform 2 enhances the transactivation activity of isoform 1 from some but not all TP53-inducible promoters. Isoform 4 suppresses transactivation activity and impairs growth suppression mediated by isoform 1. Isoform 7 inhibits isoform 1-mediated apoptosis. TP53 regulates the circadian clock by repressing CLOCK-ARNTL/BMAL1-mediated transcriptional activation of PER2.
Phospho-TP53 (S33) Recombinant Monoclonal Antibody is a recombinant antibody specifically designed to recognize the human tumor protein p53 (TP53) when it is phosphorylated at the serine 33 position. The antibody is produced by extracting antibody genes from rabbits immunized with a synthesized peptide derived from the human TP53 protein phosphorylated at S33. These genes are then introduced into expression vectors, transfected into host suspension cells, and the antibodies are produced and purified using affinity chromatography techniques .
The specificity of the antibody is validated through multiple testing methods including ELISA, immunohistochemistry (IHC), and immunofluorescence (IF) to confirm its ability to specifically interact with human TP53 protein phosphorylated at the S33 site .
Based on manufacturer recommendations and published protocols, the Phospho-TP53 (S33) Recombinant Monoclonal Antibody can be used in multiple applications with the following suggested dilutions:
Application | Recommended Dilution |
---|---|
Immunohistochemistry (IHC) | 1:50-1:200 |
Immunofluorescence (IF) | 1:20-1:200 |
Western Blotting (WB) | 1:1000 |
Proximity Ligation Assay | As specified in protocol |
These dilutions should be optimized for specific experimental conditions, including cell/tissue type, fixation method, and detection system .
Positive controls:
UV-irradiated cells (25 J/m²), harvested 4-8 hours post-treatment, as UV exposure induces significant phosphorylation at S33
Cells treated with p38 MAPK activators (e.g., anisomycin)
Cell lines with known high levels of p53 S33 phosphorylation
Negative controls:
Cell lines with p53 S33A mutations or p53-null cells (e.g., H1299)
Samples treated with lambda phosphatase to remove phosphorylation
Blocking peptide competition assays to confirm antibody specificity
p38 kinase inhibitor-treated cells, as p38 inhibition decreases phosphorylation at Ser33
For validating specificity in Western blotting, it's recommended to run phosphorylated and non-phosphorylated peptide controls alongside your experimental samples.
Phosphorylation of p53 at S33 is primarily regulated by p38 mitogen-activated protein kinase (p38 MAPK). Research has demonstrated that p38 kinase can directly phosphorylate p53 at Ser33 and Ser46 both in vitro and in vivo . The regulation of this phosphorylation is complex and context-dependent:
DNA damage response: UV radiation significantly increases p38 kinase activity, leading to enhanced phosphorylation of p53 at Ser33
Signaling pathways: Components of the Ras signaling pathway contribute to activation of p38 kinase, which subsequently phosphorylates p53 at Ser33
Protein-protein interactions: p53 and p38 kinase exist in the same physical complex, facilitating efficient phosphorylation
Kinase specificity: While p38 directly phosphorylates Ser33 and Ser46, it indirectly influences phosphorylation at other sites including Ser15 and Ser37 through a coordinated phosphorylation cascade
Experimental evidence from cell culture studies shows that inhibition of p38 activation after UV irradiation significantly decreases phosphorylation at Ser33, confirming the crucial role of this kinase in regulating this specific modification .
Phosphorylation of p53 at Ser33 plays critical roles in coordinating DNA repair, cell cycle regulation, and cell fate decisions in response to stress and damage . Key functional consequences include:
Apoptotic regulation: Mutation studies replacing Ser33 with alanine (S33A) show decreased p53-mediated and UV-induced apoptosis, indicating its importance in programmed cell death pathways
Coordinated phosphorylation: Ser33 phosphorylation appears to be a prerequisite for proper phosphorylation at other sites. Mutation of Ser33 completely blocks UV-induced phosphorylation at Ser37 and significantly decreases Ser15 phosphorylation, suggesting it functions as part of an integrated regulatory network
Transcriptional activation: While single S33A mutation shows minimal effect on p53-dependent transcription, the double mutant S33,46A exhibits significantly reduced transactivation potential, suggesting a synergistic role with other phosphorylation sites
Protein-protein interactions: Phosphorylation at Ser33 contributes to the binding energy for interactions between the p53 transactivation domain (TAD) and CREB-binding protein (CBP) domains, enhancing transcriptional coactivator recruitment
Immune system activation: Phosphorylated p53 peptides containing phospho-S33 can bind to multiple HLA-DR molecules and induce T helper responses against tumor cells expressing the phosphorylated p53 protein
Research using phospho-specific antibodies has demonstrated that under normal conditions, there is a basal level of Ser33 phosphorylation that increases modestly following UV irradiation, suggesting its involvement in both normal cellular functions and stress responses .
Phosphorylation at Ser33 participates in a complex network of post-translational modifications (PTMs) that collectively regulate p53 function:
Interdependence with other phosphorylation sites: Studies using site-specific mutants reveal that Ser33 phosphorylation is required for efficient phosphorylation at Ser37, which is completely abrogated when Ser33 is mutated to alanine. Additionally, the S33,46A double mutant shows dramatically reduced phosphorylation at Ser15
Additive contributions to binding energy: Research demonstrates that successive phosphorylation events, including at Ser33, contribute in an additive manner to the free energy for binding of the p53 transactivation domain (TAD) to the CREB-binding protein (CBP) domains
Phosphorylation cascade model: Evidence supports a model where p38 kinase-mediated phosphorylation at Ser33 initiates a phosphorylation cascade that includes Ser37 and Ser15, sites that can be phosphorylated by DNA-PK, ATM and ATR. This suggests a coordinated regulation of N-terminal phosphorylation centered on the phosphorylation at Ser33 and Ser46 by p38 kinase
Multi-kinase complexes: The interdependence of these modifications supports the hypothesis that a coordinated phosphorylation of p53 N-terminal sites may result from an association with a complex containing several kinases, rather than independent modification events
The complete understanding of these interactions requires specialized techniques such as mass spectrometry-based PTM mapping and combination of phospho-specific antibodies to detect multiple modifications simultaneously.
Several experimental approaches can be employed to specifically study S33 phosphorylation:
Site-directed mutagenesis: Creating S33A (phospho-deficient) or S33D/S33E (phospho-mimetic) mutants allows investigation of the functional consequences of phosphorylation at this site. These mutants can be expressed in p53-null cells to study specific effects
Kinase manipulation:
Phosphopeptide generation:
Detection methods:
Binding studies:
These methodologies can be combined to provide comprehensive insights into the mechanisms and functions of S33 phosphorylation in various cellular contexts.
Different cellular stressors induce distinct patterns of p53 phosphorylation at Ser33:
UV radiation: Induces significant phosphorylation at Ser33, with peak levels typically observed 4-8 hours post-treatment. This response is primarily mediated by activation of p38 MAPK
Ionizing radiation (IR): May induce different phosphorylation patterns compared to UV, often involving ATM and ATR kinases more prominently than p38 MAPK
Oxidative stress: Reactive oxygen species can activate p38 MAPK, potentially leading to increased Ser33 phosphorylation
Chemotherapeutic agents: Many DNA-damaging agents enhance the responses of CD4 T cells specific for phosphorylated p53 peptides by upregulating phosphorylated p53 expression, including at Ser33
The temporal dynamics of Ser33 phosphorylation vary depending on the stressor, with some causing rapid and transient phosphorylation while others induce more sustained modifications. These differences likely contribute to the specificity of p53-mediated responses to different types of cellular damage.
To experimentally study these changes, time-course experiments using specific stressors followed by Western blotting with phospho-specific antibodies or mass spectrometry analysis are recommended approaches.
Phosphorylated p53 peptides, including those containing phospho-Ser33, have shown promising results in tumor immunology with potential applications in immunotherapy:
T helper cell responses: The p53 22-41/Phospho-S33 peptide can induce T helper responses against tumor cells expressing the phosphorylated p53 protein. These T helper lymphocytes specifically recognize and respond to the phosphorylated epitope but not to the non-phosphorylated wild-type p53 peptide
HLA binding and population coverage: Phosphorylated p53 peptides can bind to multiple HLA-DR molecules, providing potential coverage for a broad population of cancer patients. This makes them attractive candidates for cancer immunotherapy approaches
Enhanced responses with chemotherapy: Chemotherapeutic agents have been shown to augment the responses of CD4 T cells specific for phosphorylated p53 by upregulating phosphorylated p53 expression, suggesting potential benefits of combination approaches
Clinical relevance: Evaluation of clinical samples from oropharyngeal squamous cell carcinoma revealed that 54% (13/24 cases) were positive for phosphorylated p53. Importantly, lymphocytes specific for phosphorylated p53 peptide epitopes were observed in head and neck squamous cell cancer (HNSCC) patients but not in healthy donors, suggesting that precursors of phosphorylated p53-reactive helper T lymphocytes exist in cancer patients
Immunogenicity differences: Research indicates that phosphorylated Ser33 might be more immunogenic than phosphorylated Ser37, as evidenced by stronger helper T lymphocyte responses to p-p53 S33 compared to p-p53 S37
These findings suggest that combining phosphorylated p53 peptides and chemotherapy could represent a novel immunological approach to treat certain cancers, particularly those with intact p53 that undergoes stress-induced phosphorylation.
Phospho-TP53 (S33) antibodies have potential applications in patient stratification and personalized cancer treatment:
Biomarker potential: Phosphorylated p53 at Ser33 could serve as a biomarker for:
Tumor progression and aggressiveness
DNA damage response pathway integrity
Potential responsiveness to therapies targeting p53 pathways or p38 MAPK signaling
Patient screening for immunotherapy: Detection of phosphorylated p53 in tumor samples could help identify patients more likely to respond to immunotherapeutic approaches targeting phospho-p53 epitopes. Research has shown that 54% of oropharyngeal squamous cell carcinoma cases were positive for phosphorylated p53
Combination therapy guidance: Since chemotherapeutic agents can upregulate phosphorylated p53 expression, measuring Ser33 phosphorylation status before and after initial treatment could help identify optimal combination strategies
Monitoring treatment response: Serial biopsies analyzed with phospho-specific antibodies could track changes in p53 phosphorylation status during treatment to evaluate pathway engagement and efficacy
Resistance mechanisms: Changes in Ser33 phosphorylation patterns might indicate development of resistance to certain therapies, particularly those targeting upstream kinases like p38 MAPK
Methodologically, this requires standardized protocols for tissue preparation, antibody validation across different sample types, and correlation with clinical outcomes to establish reliable cutoffs for positivity and clinical decision-making.
The phosphorylation status of p53 at Ser33, particularly in combination with other modifications, appears to influence the balance between cell cycle arrest and apoptotic responses:
Apoptosis regulation: Mutation studies replacing both Ser33 and Ser46 with alanine (S33,46A double mutant) show significantly decreased ability to induce apoptosis compared to wild-type p53. This is evidenced by:
Threshold effect: Single mutants (S33A or S46A alone) were able to suppress colony formation to the same extent as wild-type p53, suggesting a potential threshold effect where both sites need to be unphosphorylated to significantly impair apoptotic function
Coordinated regulation: The substantial reduction in apoptosis in the S33,46A double mutant correlates with:
Complete abrogation of UV-induced phosphorylation on Ser37
Significant decrease in Ser15 phosphorylation
This suggests Ser33 phosphorylation influences apoptotic responses both directly and through its effects on other phosphorylation events
Protein-protein interactions: Phosphorylation at Ser33 contributes to the binding energy for interactions between the p53 transactivation domain and transcriptional coactivators like CBP, potentially influencing the spectrum of genes activated and thus cell fate decisions
These findings suggest that phosphorylation at Ser33 plays a complex role in determining cell fate, with its importance being most evident when considered as part of the broader phosphorylation network rather than in isolation.
When encountering problems with Phospho-TP53 (S33) antibodies, researchers can implement the following troubleshooting strategies:
For non-specific binding:
Optimize blocking conditions:
Test different blocking agents (BSA, non-fat dry milk, casein, commercial blockers)
Increase blocking time and/or concentration
Include 0.1-0.3% Tween-20 in wash and antibody diluent buffers
Adjust antibody dilution:
Implement peptide competition:
Pre-incubate antibody with phosphorylated peptide to confirm specificity
Compare to non-phosphorylated peptide control to verify phospho-specificity
Use appropriate controls:
Include p53-null or S33A mutant samples
Treat half of your sample with phosphatase before detection
For weak signal issues:
Sample preparation:
Add phosphatase inhibitors (sodium fluoride, sodium orthovanadate, etc.) to all buffers
Minimize time between sample collection and processing
Consider using phospho-enrichment techniques
Enhance phosphorylation signal:
Detection enhancement:
Try signal amplification systems (e.g., biotin-streptavidin, tyramide)
For Western blotting, increase exposure time or use more sensitive substrate
For IHC/IF, optimize antigen retrieval methods (test different pH buffers)
Storage and handling:
By systematically addressing these factors, researchers can optimize their experimental conditions to achieve specific and sensitive detection of phosphorylated p53 at Ser33.
Proximity Ligation Assay (PLA) offers a powerful approach for detecting phosphorylated p53 at Ser33 with single-molecule resolution in situ. This technique is particularly valuable for studying low-abundance modifications in their native cellular context:
Principle of the assay:
The PLA requires two antibodies that bind in close proximity (< 40 nm). For phospho-TP53 (S33) detection, researchers use:
A rabbit polyclonal antibody recognizing the phospho-S33 epitope
A mouse monoclonal antibody targeting total p53
When these antibodies bind to the same p53 molecule, oligonucleotide-conjugated secondary antibodies enable amplification and detection of a fluorescent signal, visualized as discrete spots where phosphorylated p53 is present
Implementation protocol:
Fix cells using paraformaldehyde (typically 4%) and permeabilize with Triton X-100
Block non-specific binding sites with appropriate blocking solution
Incubate with primary antibodies (rabbit anti-phospho-TP53 S33 at 1:1200 dilution and mouse anti-TP53 at 1:50 dilution)
Add PLA probes (secondary antibodies with attached oligonucleotides)
Perform ligation and amplification steps according to manufacturer's protocol
Counterstain nuclei with DAPI and image using fluorescence microscopy
Analysis and quantification:
Advantages over traditional methods:
Single-molecule sensitivity
Visualization of spatial distribution within cells
Elimination of non-specific background through requirement for dual antibody binding
Quantitative assessment of phosphorylation events in situ
This approach is particularly valuable for studying the dynamics of p53 phosphorylation in response to different stressors, in different subcellular compartments, or in heterogeneous cell populations within tissue samples.
The use of phosphorylated p53 epitopes for cancer immunotherapy represents an emerging approach with several promising aspects:
Generation of phospho-epitope specific T cells:
Synergy with conventional cancer treatments:
Experimental protocols for T cell generation:
Isolate peripheral blood mononuclear cells (PBMCs) from cancer patients or healthy donors
Stimulate with synthetic phosphorylated p53 peptides (including p53 22-41/Phospho-S33)
Expand T cells using appropriate cytokines (IL-2, IL-7, etc.)
Test specificity using peptide-pulsed APCs and measure responses via cytokine production, proliferation, or cytotoxicity assays
Clinical potential:
Analysis of head and neck squamous cell carcinoma (HNSCC) patients showed pre-existing T cell responses to phosphorylated p53 peptides, indicating natural immunogenicity
54% of oropharyngeal squamous cell carcinoma samples were positive for phosphorylated p53, representing a substantial patient population that might benefit from this approach
MHC coverage and population applicability:
These findings point to a novel immunotherapeutic strategy that targets tumor-specific post-translational modifications rather than tumor-specific proteins, potentially offering greater specificity with reduced off-target effects against normal tissues.
The interconnection between p38 MAPK signaling, p53 Ser33 phosphorylation, and therapeutic response in cancer represents an important area for translational research:
Signaling pathway integration:
p38 MAPK directly phosphorylates p53 at Ser33 and Ser46, forming a critical link between cellular stress signaling and p53 activation
Components of the Ras signaling pathway contribute to activation of p38 kinase and subsequent p53 phosphorylation, creating cross-talk between oncogenic and tumor suppressor pathways
p53 and p38 kinase exist in the same physical complex, facilitating efficient signal transduction
Impact on therapeutic responses:
Inhibition of p38 activation after UV irradiation decreases phosphorylation at Ser33, Ser37, and Ser15, and markedly reduces UV-induced apoptosis in a p53-dependent manner
This suggests that integrity of the p38-p53 phosphorylation axis may be required for optimal response to DNA-damaging therapies
Conversely, upregulation of this pathway might sensitize resistant tumors to conventional treatments
Experimental evidence from treatment models:
Potential therapeutic approaches:
Activation strategy: Enhancing p38 MAPK activity in tumors with wild-type p53 could increase p53 phosphorylation and promote apoptosis
Inhibition strategy: In contexts where p53 is mutated or has oncogenic functions, blocking p38-mediated phosphorylation might provide therapeutic benefit
Immunotherapeutic approach: Using phospho-S33 peptide vaccination in combination with agents that increase p38 activity and p53 phosphorylation
Biomarker applications:
Phospho-S33 status could serve as a biomarker for:
Functional p38-p53 signaling axis
Potential responsiveness to DNA-damaging therapies
Selection of patients for combination treatments targeting this pathway
The intricate relationship between these pathways offers multiple points for therapeutic intervention and suggests that personalized approaches based on the status of p38-p53 signaling could improve treatment outcomes in various cancer types.