TP53I3 Human

Tumor Protein p53 Inducible Protein 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to TP53I3 Human

TP53I3 (Tumor Protein p53 Inducible Protein 3), also known as PIG3, is a human recombinant protein encoded by the TP53I3 gene. It is a p53-inducible oxidoreductase implicated in DNA damage response, reactive oxygen species (ROS) regulation, and apoptosis . TP53I3 is synthesized as a 37.6 kDa polypeptide chain containing 352 amino acids, including a 20-amino acid His tag at the N-terminus for purification . Its structural and functional roles in cancer biology make it a critical focus of oncology research.

Key Features:

PropertyDetails
Molecular Weight37.6 kDa
Amino Acid Sequence352 residues (1–332 a.a. native + 20 His-tag)
Expression SystemEscherichia coli
PurificationStandard chromatography
Formulation20 mM Tris HCl (pH 8), 0.1 M NaCl, 10% glycerol
StabilityStore at 4°C (short-term) or -20°C (long-term); carrier protein recommended for extended storage

Biological Functions and Mechanisms

TP53I3 is transcriptionally activated by the tumor suppressor p53, which binds to a pentanucleotide microsatellite sequence in its promoter . Key roles include:

  • Apoptosis Induction: Mediates p53-dependent apoptosis via ROS generation and mitochondrial membrane destabilization .

  • DNA Damage Response: Facilitates homologous recombination repair (HRR) of double-strand breaks (DSBs) .

  • Oxidative Stress Regulation: Catalyzes quinone redox reactions, influencing cellular responses to oxidative damage .

Mechanism in Cancer:

ProcessImpact of TP53I3
ROS ProductionGenerates superoxide radicals, triggering apoptosis in damaged cells .
HRR EfficiencyKnockdown reduces HRR by ~20%, increasing genomic instability .
Chemotherapy ResponseTP53I3-deficient cells show hypersensitivity to bleomycin, mitomycin C, and etoposide .

Clinical Associations:

Cancer TypeRole of TP53I3References
Ovarian CancerGermline truncating mutations (e.g., p.S252X) linked to impaired HRR and chemoresistance .
Non-Small Cell Lung CancerOverexpression correlates with poor prognosis .
Breast CancerPolymorphisms in TP53I3’s microsatellite region influence cancer susceptibility .

Experimental Insights:

  • TP53I3 p.S252X mutation reduces ROS formation under oxidative stress, conferring resistance to etoposide and hydrogen peroxide .

  • Loss of TP53I3 expression promotes tumor progression in colon and thyroid cancers .

Therapeutic Implications

TP53I3’s role in p53-mediated apoptosis and DNA repair positions it as a potential target for:

  • Chemotherapy Sensitization: Restoring TP53I3 function may enhance drug efficacy in TP53-mutant cancers .

  • Synthetic Lethality Strategies: Targeting TP53I3-deficient cancers with PARP inhibitors or ROS-inducing agents .

Challenges and Future Directions

Despite its therapeutic potential, challenges include:

  • Complex Regulation: TP53I3’s activity depends on p53 status and ROS dynamics, complicating targeted interventions .

  • Mutation Variability: Polymorphisms in its microsatellite region influence transcriptional activation, requiring personalized approaches .

Product Specs

Introduction
TP53I3 is involved in the production of reactive oxygen species (ROS). It exhibits low NADPH-dependent naphtoquinone reductase activity, favoring 1,2-naphtoquinone over 1,4-naphtoquinone. Additionally, TP53I3 displays weak NADPH-dependent diamine reductase activity in vitro. Found in the cytoplasm, TP53I3 is upregulated in both primary and transformed cell cultures upon exposure to genotoxic agents. Notably, microsatellite polymorphism in TP53I3 is linked to varying levels of susceptibility to cancer.
Description
Recombinant Human TP53I3, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 352 amino acids (residues 1-332) with a molecular weight of 37.6 kDa. A 20 amino acid His tag is fused to the N-terminus of the TP53I3 protein. Purification is achieved through standard chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The solution contains 1mg/ml of TP53I3 in a buffer consisting of 20mM Tris HCl (pH 8), 0.1M NaCl, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the solution should be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is advisable for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined to be greater than 90% using SDS-PAGE analysis.
Synonyms
TP53I3, PIG3, Quinone Oxidoreductase, tumor protein p53 inducible protein 3.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MLAVHFDKPG GPENLYVKEV AKPSPGEGEV LLKVAASALN RADLMQRQGQ YDPPPGASNI LGLEASGHVAELGPGCQGHW KIGDTAMALL PGGGQAQYVT VPEGLLMPIP EGLTLTQAAA IPEAWLTAFQ LLHLVGNVQA GDYVLIHAGL SGVGTAAIQLTRMAGAIPLV TAGSQKKLQM AEKLGAAAGF NYKKEDFSEA TLKFTKGAGV NLILDCIGGS YWEKNVNCLA LDGRWVLYGL MGGGDINGPLFSKLLFKRGS LITSLLRSRD NKYKQMLVNA FTEQILPHFS TEGPQRLLPV LDRIYPVTEI QEAHKYMEAN KNIGKIVLEL PQ.

Q&A

What is TP53I3 and what is its primary function in human cells?

TP53I3 (Tumor Protein P53 Inducible Protein 3) is a gene that is transcriptionally activated by the tumor suppressor protein p53. It plays a crucial role in cellular processes related to DNA damage response and reactive oxygen species (ROS)-induced apoptosis . The protein encoded by this gene is believed to function as part of the p53-mediated cellular response to various stress conditions. TP53I3 is involved in quinone oxidoreductase activity, which contributes to its role in managing oxidative stress within cells. It serves as an important downstream effector in the p53 pathway, which is central to tumor suppression mechanisms through regulation of cell cycle arrest, DNA repair, and programmed cell death.

How is TP53I3 expression regulated in normal and cancer cells?

TP53I3 expression is primarily regulated by p53, which binds to the promoter region of TP53I3 and increases its transcription in response to cellular stress or DNA damage . In normal cells, TP53I3 expression is typically maintained at basal levels but becomes upregulated when p53 is activated by various stress stimuli. In cancer cells, TP53I3 regulation can be significantly altered depending on the p53 status of the cells.

In cells with wild-type p53, various interventions that activate p53 can lead to increased TP53I3 expression. For example, research has shown that targeting long non-coding RNAs (lncRNAs) such as MEG3 and linc-ROR can significantly upregulate TP53I3 expression in colorectal cancer cells. In HCT116 cells, this intervention increased TP53I3 transcript levels by 3.36-fold, while in SW480 cells, an even more dramatic increase of 8.67-fold was observed . Conversely, in cells with mutant p53 or p53 deficiency, TP53I3 expression is often dysregulated, contributing to cancer progression.

What are the known mutations in TP53I3 and their clinical significance?

Several mutations in TP53I3 have been identified, with varying clinical significance in cancer development and progression. One notably studied variant is the p.S252X truncating mutation, which has been associated with decreased homologous recombination repair (HRR) efficiency . This mutation creates a premature stop codon that results in a truncated protein with impaired function.

While germline variants in TP53I3 associated with cancer risk are still being investigated, studies have identified truncating mutations in TP53I3 in hereditary breast and ovarian cancer (HBOC) patients, suggesting its potential role in cancer predisposition .

What experimental models are commonly used to study TP53I3 function?

Researchers employ various experimental models to investigate TP53I3 function, each with specific advantages for addressing different research questions:

Cell Line Models:

  • HeLa-DR-GFP cells: Used to study the role of TP53I3 in homologous recombination repair through GFP expression as a proxy for HRR efficiency

  • HCT116 and SW480 colorectal cancer cell lines: Commonly used to study TP53I3's role in colorectal cancer progression and its response to various interventions

Experimental Approaches:

  • siRNA Knockdown: Utilized to reduce TP53I3 expression and observe the resulting phenotypic changes in DNA repair capacity, cell viability, and drug sensitivity

  • Plasmid Rescue Experiments: Wild-type and mutant TP53I3 plasmids are transfected into cells after siRNA knockdown to assess functional rescue

  • Drug Sensitivity Assays: Cells with manipulated TP53I3 expression are treated with chemotherapeutic agents like bleomycin, mitomycin C, and etoposide to evaluate drug sensitivity

  • Vector-based Expression Systems: Bidirectional and unidirectional vectors containing sequences that affect TP53I3 expression are used to study its involvement in cancer cell proliferation

These experimental models allow researchers to investigate TP53I3's functional roles in various cellular processes and its contribution to cancer development and treatment response.

How does TP53I3 contribute to DNA damage response and repair mechanisms?

TP53I3 plays a significant role in DNA damage response and repair mechanisms, particularly in homologous recombination repair (HRR) of double-strand breaks (DSBs). Experimental evidence has demonstrated that knockdown of TP53I3 with siRNA results in a significant reduction in HRR capabilities by approximately 20% (p ≤ 0.01) compared to control conditions . This places TP53I3 in the company of well-established DNA repair genes like ATM, BRCA1, BRCA2, CHEK2, PALB2, RAD51D, and RAD50, all of which are associated with the HRR mechanism.

When examining the specific effects of TP53I3 mutations, research has shown that the p.S252X truncation variant significantly impairs HRR activity. In rescue experiments, transfection with wild-type TP53I3 successfully restored HRR function after siRNA knockdown, whereas the truncated TP53I3-S252X mutant failed to restore normal HRR levels . This finding indicates that the C-terminal portion of the protein, which is lost in the p.S252X variant, is critical for its DNA repair function.

The mechanism by which TP53I3 contributes to DNA repair appears to be distinct from its role in p53-mediated apoptosis. Unlike TP53AIP1, another p53-inducible gene primarily involved in apoptosis, TP53I3 has a direct functional impact on the cell's ability to repair DNA damage. This dual functionality makes TP53I3 a particularly important player in the cellular response to genotoxic stress, influencing both repair and apoptotic pathways.

What is the role of TP53I3 in chemosensitivity and potential applications in cancer treatment?

TP53I3 significantly influences cellular sensitivity to various chemotherapeutic agents, making it a potential target for enhancing cancer treatment efficacy. Research has demonstrated that depletion of TP53I3 renders cells more sensitive to several DNA-damaging agents, including bleomycin, mitomycin C (MMC), and etoposide .

Chemosensitivity Data:

Treatment ConditionCell Viability (%)
Control (scramble)~80%
TP53I3 knockdown alone~60% (p ≤ 0.001)
TP53I3 knockdown + wt-TP53I3 rescue~80%
TP53I3 knockdown + TP53I3-S252X rescue~80%

When cells with TP53I3 knockdown were treated with chemotherapeutic agents at their IC50 dosages (100 nM for mitomycin C, 1.5 μM for bleomycin, and 4 μM for etoposide), they exhibited significantly enhanced sensitivity compared to control cells . This suggests that TP53I3 status could be a determinant of treatment response in cancer patients.

The potential applications in cancer treatment are twofold:

  • Predictive Biomarker: TP53I3 expression or mutation status could serve as a biomarker to predict patient response to specific chemotherapeutic regimens, particularly those involving DNA-damaging agents.

  • Therapeutic Target: Modulating TP53I3 expression or activity in combination with conventional chemotherapy might enhance treatment efficacy. For instance, in colorectal cancer, simultaneous targeting of lncRNAs that affect TP53I3 expression has shown promising results in inhibiting cancer cell proliferation .

Interestingly, the relationship between TP53I3 and chemosensitivity appears to be complex. While TP53I3 knockdown increases sensitivity to chemotherapeutic agents, rescue with either wild-type or mutant TP53I3 restores cell viability to control levels . This suggests that the role of TP53I3 in drug response may involve mechanisms beyond its function in DNA repair, possibly including effects on apoptosis or cell cycle regulation.

How does TP53I3 interact with reactive oxygen species (ROS) in cellular stress responses?

TP53I3 plays a critical role in the cellular response to oxidative stress through its interaction with reactive oxygen species (ROS). As a quinone oxidoreductase, TP53I3 is directly involved in ROS metabolism, which is particularly relevant in cancer cells that often exhibit altered redox states.

Research has demonstrated that TP53I3 influences ROS formation under oxidative stress conditions. When cells expressing the truncated TP53I3 p.S252X variant were exposed to oxidative stress from hydrogen peroxide or etoposide, they exhibited a reduction in ROS formation compared to cells with wild-type TP53I3 . This finding is significant because ROS are important precursors to apoptosis, suggesting that TP53I3 mutations might affect cancer cell survival by altering ROS-mediated cell death pathways.

The interaction between TP53I3 and ROS appears to be bidirectional:

  • TP53I3 as a ROS Regulator: Through its enzymatic activity, TP53I3 can influence cellular ROS levels, potentially affecting oxidative DNA damage and subsequent repair processes.

  • ROS as TP53I3 Activators: Oxidative stress can trigger p53 activation, which in turn upregulates TP53I3 expression, creating a feedback loop in the cellular stress response.

What is the relationship between TP53I3 and other p53 target genes in cancer progression?

TP53I3 functions within a complex network of p53 target genes that collectively regulate cell fate decisions in response to various stressors. Understanding the interactions between TP53I3 and other p53-regulated genes provides insights into the broader p53 pathway dynamics in cancer progression.

Research in colorectal cancer has demonstrated coordinated regulation of several p53 target genes when TP53I3 expression is modulated. When TP53 is activated through interventions targeting lncRNAs (MEG3 and linc-ROR), there is a concurrent upregulation of multiple p53 target genes including TP53INP1, TP53I3, GDF15, CDKN1A (p21), and BAX, accompanied by downregulation of cell cycle regulators like CCND1 (cyclin D1) and CCNE1 (cyclin E1) .

Comparative Expression Changes in Response to p53 Activation:

GeneFold Change in HCT116 CellsFold Change in SW480 Cells
TP5310.436.16
TP53INP12.268.48
TP53I33.368.67
GDF152.031.77
CDKN1A2.242.67
BAX2.311.78
CCND10.690.59
CCNE10.440.45

This coordinated expression pattern reveals that TP53I3 functions as part of a broader p53-mediated transcriptional program that regulates multiple cellular processes including cell cycle arrest (through CDKN1A), apoptosis (through BAX), and other tumor-suppressive mechanisms.

Interestingly, the relationship between TP53I3 and other p53 target genes appears to be cell-type specific. The expression changes in HCT116 and SW480 colorectal cancer cells show different magnitudes, indicating that the p53 pathway is modulated by cellular context . This variability may explain why TP53I3 can function as either a tumor suppressor or promoter depending on the cancer type.

The interplay between TP53I3 and genes like TP53INP1 is particularly noteworthy, as both are involved in p53-mediated responses but appear to have distinct functions. While TP53I3 affects both DNA repair and ROS-mediated apoptosis, TP53INP1 is primarily associated with p53-dependent apoptosis . This functional specialization within the p53 network allows for nuanced cellular responses to different types and levels of stress.

What techniques are most effective for measuring TP53I3 expression and activity in different tissue types?

Accurately measuring TP53I3 expression and activity across different tissue types requires a multi-faceted approach that addresses both transcript and protein levels, as well as functional assessments. The following methodologies have proven most effective in current research:

RNA Expression Analysis:

  • Quantitative Real-Time PCR (qRT-PCR): This technique has been successfully employed to measure TP53I3 transcript levels in colorectal cancer cell lines (HCT116 and SW480) and is particularly useful for detecting time-dependent changes in expression . The high sensitivity of qRT-PCR makes it suitable for detecting small changes in TP53I3 expression across different experimental conditions.

  • RNA Sequencing (RNA-seq): For broader transcriptome analysis, RNA-seq provides comprehensive insights into TP53I3 expression patterns in relation to other genes in the p53 pathway, allowing for identification of co-expression networks and novel regulatory relationships.

Protein Detection Methods:

  • Western Blotting: Western blot analysis is essential for confirming that changes in transcript levels translate to altered protein expression. This technique has been used to validate TP53I3 protein levels after siRNA knockdown and plasmid rescue experiments .

  • Immunohistochemistry (IHC): For tissue-specific analysis, particularly in patient samples, IHC allows visualization of TP53I3 protein distribution and expression levels within the tissue microenvironment, providing spatial context that other methods lack.

Functional Assays:

  • Homologous Recombination Repair (HRR) Assay: The HeLa-DR-GFP reporter system has been effectively used to assess TP53I3's functional impact on DNA repair, where GFP expression serves as a proxy for HRR efficiency . This assay is particularly valuable for evaluating the functional consequences of TP53I3 variants.

  • ROS Detection Assays: Fluorescent probes for detecting reactive oxygen species are useful for measuring TP53I3's impact on oxidative stress responses, particularly when comparing wild-type and mutant variants under stress conditions.

  • Cell Viability and Drug Sensitivity Tests: Colony formation assays provide quantitative assessment of TP53I3's influence on cell survival, especially in response to chemotherapeutic agents . These functional readouts are crucial for understanding the biological significance of altered TP53I3 expression.

When studying TP53I3 across different tissue types, combining these methodologies provides the most comprehensive assessment. Tissue-specific considerations include optimizing extraction protocols for different sample types and accounting for potential confounding factors such as tissue heterogeneity and variable p53 status across different cancer types.

How can researchers effectively model TP53I3 mutations to study their impact on cancer development?

Effectively modeling TP53I3 mutations requires a multi-tiered approach that spans computational prediction, in vitro functional validation, and in vivo systems. The following methodological framework has proven valuable for comprehensive investigation of TP53I3 variants:

Computational Approaches:

  • In Silico Prediction Tools: Utilizing algorithms like SIFT, PolyPhen-2, and MutationTaster to predict the functional impact of TP53I3 variants based on evolutionary conservation and protein structure. These computational tools provide initial screening to prioritize variants for experimental validation .

  • Structural Modeling: Employing protein structure prediction software to model how specific mutations (like p.S252X) might affect TP53I3 protein folding, stability, and functional domains. This approach helps generate hypotheses about mechanism-specific effects of mutations.

In Vitro Cellular Models:

  • Site-Directed Mutagenesis: Creating precise mutations in TP53I3 expression constructs to generate wild-type and mutant versions for functional comparison. This technique was successfully used to create the TP53I3 p.S252X variant for functional studies .

  • Rescue Experiments: Knocking down endogenous TP53I3 with siRNA followed by transfection with either wild-type or mutant TP53I3 constructs. This approach effectively demonstrates whether mutant variants retain functional capacity. Research has shown that while wild-type TP53I3 successfully rescued HRR function, the p.S252X variant failed to do so .

  • CRISPR/Cas9 Gene Editing: Introducing specific TP53I3 mutations into relevant cell lines to create isogenic models that differ only in TP53I3 status. This approach minimizes confounding factors associated with different genetic backgrounds.

Functional Validation Assays:

  • DNA Repair Efficiency: Using reporter systems like HeLa-DR-GFP to quantify homologous recombination repair capacity in cells expressing different TP53I3 variants .

  • Chemosensitivity Testing: Exposing cells with different TP53I3 variants to DNA-damaging agents to assess their influence on drug response. This approach has revealed that TP53I3 status affects sensitivity to bleomycin, mitomycin C, and etoposide .

  • ROS Formation Assays: Measuring reactive oxygen species levels in response to oxidative stress inducers to evaluate how TP53I3 mutations affect this important precursor to apoptosis .

In Vivo Models:

  • Patient-Derived Xenografts (PDXs): Establishing xenograft models from patient tumors with different TP53I3 variants to study their behavior in a more physiologically relevant context.

  • Genetically Engineered Mouse Models (GEMMs): Creating mice with specific TP53I3 mutations to evaluate their impact on cancer initiation, progression, and treatment response.

The combination of in silico and wet laboratory approaches has proven effective in evaluating variants of uncertain significance (VUSs) in TP53I3 and establishing novel germline predisposition genetic loci . This integrated approach provides a more comprehensive understanding of how TP53I3 mutations contribute to cancer development and progression.

What are the best experimental designs for investigating TP53I3's role in chemoresistance mechanisms?

Investigating TP53I3's role in chemoresistance requires carefully designed experiments that address both mechanistic questions and clinical relevance. The following experimental approaches are recommended for comprehensive assessment:

Baseline Expression and Correlation Studies:

  • Expression Profiling Across Cancer Cell Lines: Systematically analyze TP53I3 expression levels across a panel of cancer cell lines with known chemoresistance profiles. Correlate expression levels with IC50 values for various chemotherapeutic agents to identify potential associations between TP53I3 and drug resistance.

  • Patient Sample Analysis: Examine TP53I3 expression in matched pre- and post-treatment tumor samples from patients who developed resistance during therapy. This approach provides clinically relevant insights into TP53I3's role in acquired resistance.

Genetic Manipulation Experiments:

  • Loss-of-Function Studies: Use siRNA or shRNA to knock down TP53I3 expression in chemosensitive cells, then assess changes in drug sensitivity. Research has demonstrated that TP53I3 knockdown significantly increases sensitivity to bleomycin, mitomycin C, and etoposide .

  • Gain-of-Function Studies: Overexpress wild-type TP53I3 in cells with low endogenous expression to determine if this confers resistance to chemotherapeutic agents.

  • Variant-Specific Effects: Compare the impact of wild-type TP53I3 versus mutant variants (like p.S252X) on chemosensitivity profiles. This approach helps distinguish between loss-of-function and gain-of-function effects of mutations .

Mechanistic Investigation:

  • DNA Damage Response Assays: Quantify DNA damage markers (e.g., γ-H2AX foci) after drug treatment in cells with manipulated TP53I3 expression to assess its influence on DNA damage response pathways.

  • Cell Death Pathway Analysis: Evaluate markers of apoptosis, necrosis, and other cell death mechanisms to determine how TP53I3 affects cell fate decisions after drug exposure.

  • ROS Measurement: Monitor reactive oxygen species levels in response to oxidative stress-inducing agents, as TP53I3 has been shown to influence ROS formation, a critical factor in the efficacy of many chemotherapeutics .

Combinatorial Approaches:

  • Drug Combination Studies: Assess whether TP53I3 modulation can enhance the efficacy of standard chemotherapeutic regimens through synergistic interactions.

  • Pathway-Specific Inhibitors: Combine TP53I3 manipulation with inhibitors of related pathways (e.g., DNA repair, apoptosis) to identify potential synthetic lethal interactions that could be exploited therapeutically.

Experimental Design Considerations:

  • Dose-Response Curves: Generate complete dose-response curves rather than testing single drug concentrations to accurately characterize shifts in drug sensitivity.

  • Time-Course Experiments: Evaluate drug responses at multiple time points to capture both immediate and delayed effects of TP53I3 on chemoresistance.

  • Multiple Cell Lines: Use diverse cell line models to account for genetic background differences that might influence TP53I3's role in drug response.

  • Clinically Relevant Drug Concentrations: Ensure that experiments utilize drug concentrations within the clinically achievable range for translational relevance.

This comprehensive experimental approach would provide robust insights into TP53I3's contribution to chemoresistance mechanisms and potential strategies for overcoming resistance in cancer therapy.

How can researchers integrate TP53I3 data with broader p53 pathway analysis in cancer genomics studies?

Integrating TP53I3 data with broader p53 pathway analysis requires sophisticated computational and experimental approaches that capture the complex interrelationships within this critical tumor suppressor network. Below is a methodological framework for comprehensive integration:

Multi-Omics Data Integration:

Network Analysis Approaches:

  • Pathway Enrichment Analysis: Use tools like GSEA (Gene Set Enrichment Analysis) to identify enriched pathways associated with TP53I3 expression changes. This approach has revealed connections between TP53I3 and other p53 target genes in colorectal cancer research .

  • Protein-Protein Interaction Networks: Map the interactions between TP53I3 and other p53 pathway proteins using databases like STRING or BioGRID, supplemented with experimental validation through co-immunoprecipitation or proximity ligation assays.

  • Gene Regulatory Networks: Construct networks that capture transcriptional regulation relationships within the p53 pathway, including feedback loops and co-regulation patterns between TP53I3 and other p53 targets.

Functional Genomics Integration:

  • CRISPR Screens: Perform genome-wide or focused CRISPR screens to identify synthetic lethal interactions with TP53I3 deficiency or mutation, placing TP53I3 function in the context of broader cellular pathways.

  • Combinatorial Perturbation Experiments: Simultaneously manipulate TP53I3 and other p53 pathway genes to assess epistatic relationships and functional redundancy within the pathway.

Clinical Data Integration:

Computational Tools and Resources:

  • Pathway Visualization Tools: Use tools like Cytoscape or PathVisio to visually represent the relationships between TP53I3 and other p53 pathway components based on experimental data.

  • Cancer Genomics Databases: Leverage resources like TCGA, ICGC, and cBioPortal to access large-scale cancer genomics data for integrative analysis of TP53I3 in the context of p53 pathway alterations across different cancer types.

  • Predictive Modeling: Develop machine learning models that incorporate TP53I3 and other p53 pathway features to predict cancer phenotypes, treatment responses, or patient outcomes.

Methodological Example:
A comprehensive approach has been demonstrated in colorectal cancer research, where interventions targeting lncRNAs led to coordinated changes in the expression of multiple p53 pathway genes, including TP53, TP53INP1, TP53I3, GDF15, CDKN1A, BAX, CCND1, and CCNE1 . This study exemplifies how manipulating one component of the pathway (p53 itself) affects the expression of multiple downstream targets, including TP53I3, in a cell type-specific manner. The differential responses observed in HCT116 and SW480 cells highlight the importance of considering cellular context when integrating TP53I3 data with broader p53 pathway analysis.

By implementing these integrative approaches, researchers can place TP53I3 within its functional context in the p53 pathway and develop a more comprehensive understanding of how alterations in this gene contribute to cancer development, progression, and treatment response.

How might TP53I3 be targeted therapeutically, and what biomarker strategies could predict response?

Developing therapeutic strategies targeting TP53I3 represents a promising avenue for cancer treatment, particularly in tumors with intact p53 function or specific TP53I3 alterations. Several potential therapeutic approaches warrant investigation:

Direct Targeting Approaches:

  • Small Molecule Activators: Design compounds that enhance TP53I3 activity, particularly in cancers where its expression is downregulated. This approach could leverage the protein's quinone oxidoreductase function to promote ROS-mediated cell death in cancer cells.

  • Gene Therapy Approaches: Develop vectors for TP53I3 restoration in tumors with reduced expression. The bidirectional vector system used in colorectal cancer research provides a potential template, where simultaneous targeting of MEG3 and linc-ROR effectively upregulated TP53I3 expression and inhibited cancer cell proliferation .

  • Synthetic Lethality Exploitation: Identify and target genes or pathways that become essential in the context of TP53I3 deficiency or mutation, similar to the PARP inhibitor approach for BRCA-deficient cancers.

Indirect Targeting Strategies:

  • p53 Pathway Modulation: Utilize compounds that activate wild-type p53 or restore mutant p53 function to indirectly increase TP53I3 expression and activity. Several p53-targeting therapies are already in clinical development for various cancers .

  • Epigenetic Modifiers: Apply epigenetic drugs like DNA methyltransferase inhibitors or histone deacetylase inhibitors to reverse epigenetic silencing of TP53I3 in cancers where this mechanism contributes to reduced expression.

  • Combination Approaches: Combine TP53I3-targeting strategies with conventional chemotherapeutics, particularly DNA-damaging agents like bleomycin, mitomycin C, and etoposide, which have shown enhanced efficacy in cells with TP53I3 deficiency .

Biomarker Strategies for Patient Selection:

  • TP53I3 Expression Profiling: Quantify TP53I3 mRNA and protein expression levels to identify patients with aberrant expression who might benefit from targeting approaches.

  • Mutation Screening: Detect specific TP53I3 mutations, such as p.S252X, that affect protein function and might predict response to certain therapies or combination approaches .

  • p53 Status Assessment: Determine p53 mutation status as a complementary biomarker, since TP53I3 is a p53 target gene and its regulation depends on functional p53.

  • DNA Repair Capacity Measurement: Develop functional assays to assess homologous recombination repair efficiency as a surrogate marker for TP53I3 activity, potentially predicting response to DNA-damaging agents.

  • Oxidative Stress Markers: Measure baseline ROS levels and oxidative stress markers in tumors to identify patients who might benefit from therapies targeting the TP53I3-ROS axis.

Companion Diagnostic Development:
To implement these biomarker strategies effectively, companion diagnostic assays would need to be developed and validated alongside therapeutic approaches. These might include:

  • Immunohistochemistry protocols for TP53I3 protein detection in tumor samples

  • Targeted sequencing panels including TP53I3 and related p53 pathway genes

  • Functional assays to measure DNA repair capacity in patient-derived samples

  • ROS detection methods adaptable to clinical samples

Research in colorectal cancer has already demonstrated the potential of targeting approaches that affect TP53I3 expression, showing significant effects on cell proliferation, apoptosis, and cell cycle arrest . The differential responses observed between HCT116 and SW480 cells highlight the importance of cellular context and the need for precision medicine approaches in targeting TP53I3.

What are the unexplored connections between TP53I3 and emerging cancer biology concepts like tumor metabolism and immunology?

TP53I3's potential involvement in tumor metabolism and immunology represents an exciting frontier in cancer research. While direct evidence is still emerging, several conceptual connections warrant exploration:

TP53I3 in Cancer Metabolism:

  • ROS Metabolism Connection: TP53I3's role in reactive oxygen species regulation directly intersects with cancer metabolism, as altered redox states are a hallmark of metabolic reprogramming in cancer cells. The enzyme's quinone oxidoreductase activity suggests it may influence NAD+/NADH ratios, which are critical for numerous metabolic pathways.

  • p53-Metabolic Axis: Recent research has established that p53 regulates multiple metabolic pathways including glycolysis, oxidative phosphorylation, and lipid metabolism . As a p53 target gene, TP53I3 may contribute to these regulatory effects, particularly in stress responses that trigger metabolic adaptation.

  • Ferroptosis Regulation: p53 is known to regulate ferroptosis, an iron-dependent form of programmed cell death characterized by lipid peroxidation . Given TP53I3's involvement in ROS metabolism, investigating its potential role in ferroptosis sensitivity or resistance could reveal novel therapeutic opportunities.

TP53I3 in Cancer Immunology:

  • Damage-Associated Molecular Patterns (DAMPs): Cell death processes influenced by TP53I3, particularly those involving ROS, may affect the release of DAMPs that modulate immune response to cancer cells. This could link TP53I3 function to immunogenic cell death, a clinically relevant concept in immunotherapy.

  • Tumor Microenvironment Modulation: ROS levels in the tumor microenvironment significantly affect immune cell function. If TP53I3 influences extracellular ROS, it may indirectly shape the immune landscape within tumors, affecting immunotherapy response.

  • Stress-Induced Ligand Expression: p53 activation can induce expression of ligands for immune receptors like NKG2D. As a p53 target, TP53I3 might be co-regulated with these immune-related genes, suggesting potential roles in immune recognition of cancer cells.

Emerging Research Directions:

  • Metabolic Phenotyping: Comprehensive metabolic profiling of cells with different TP53I3 expression levels or mutations could reveal specific metabolic pathways affected by this gene.

  • Immune Infiltrate Correlation: Analyzing the relationship between TP53I3 expression in tumors and patterns of immune cell infiltration could identify previously unrecognized associations with the immune microenvironment.

  • Extracellular Vesicle Content: Investigating whether TP53I3 expression affects the content or release of extracellular vesicles could reveal novel mechanisms of intercellular communication relevant to both metabolism and immunity.

  • Immunotherapy Response Prediction: Evaluating whether TP53I3 status correlates with response to immune checkpoint inhibitors or other immunotherapies could identify it as a potential biomarker for treatment selection.

  • Metabolic-Immune Intersection: Exploring how TP53I3-mediated metabolic changes might influence immune cell function in the tumor microenvironment could reveal novel therapeutic strategies combining metabolic and immune targeting.

These unexplored connections represent promising avenues for future research that could significantly expand our understanding of TP53I3's role in cancer biology beyond its established functions in DNA repair and apoptosis. Given the growing importance of both metabolism and immunology in cancer treatment, elucidating TP53I3's involvement in these areas could lead to innovative therapeutic approaches.

What methodological advances are needed to better understand the tissue-specific functions of TP53I3?

Advancing our understanding of TP53I3's tissue-specific functions requires innovative methodological approaches that address the complexity of gene regulation and protein function across diverse cellular contexts. The following methodological advances would significantly enhance research in this area:

Advanced 3D Culture Systems:

  • Organoid Technology: Develop tissue-specific organoid models from different organs with controlled TP53I3 expression or mutations. These models would better recapitulate the native tissue architecture and cellular heterogeneity compared to traditional 2D cultures.

  • Tissue-on-a-Chip Platforms: Create microfluidic devices that mimic tissue-specific microenvironments to study TP53I3 function under physiologically relevant conditions, including appropriate extracellular matrix components and mechanical forces.

  • Co-Culture Systems: Establish co-culture models incorporating multiple cell types from the same tissue to investigate how TP53I3 function in one cell type affects neighboring cells, addressing the complexity of tissue-specific responses.

Single-Cell Analysis Techniques:

  • Single-Cell RNA Sequencing: Apply scRNA-seq to tissues or heterogeneous cell populations to reveal cell type-specific expression patterns of TP53I3 and associated gene networks, providing insights into differential regulation across cell types within the same tissue.

  • Single-Cell Proteomics: Develop methods for analyzing TP53I3 protein levels and post-translational modifications at the single-cell level to understand cell-to-cell variability in protein function.

  • Spatial Transcriptomics: Utilize techniques that preserve spatial information while measuring gene expression to understand how TP53I3 expression varies across different regions of a tissue or tumor.

Tissue-Specific Genetic Modeling:

  • Conditional Knockout/Knockin Models: Create tissue-specific TP53I3 knockout or mutant knockin animal models using Cre-lox or similar systems to evaluate phenotypic consequences in a tissue-selective manner.

  • Inducible Expression Systems: Develop tissue-specific inducible expression systems for wild-type and mutant TP53I3 to study temporal aspects of gene function during development or disease progression.

  • Humanized Mouse Models: Generate humanized mouse models with human TP53I3 variants to better model human disease in a physiologically relevant context.

Multi-Omics Integration at Tissue Level:

  • Tissue-Specific Epigenomic Profiling: Characterize tissue-specific epigenetic landscapes (chromatin accessibility, histone modifications, DNA methylation) around the TP53I3 locus to understand differential regulation mechanisms.

  • Proteome-Transcriptome Correlation: Develop analytical frameworks to correlate TP53I3 transcript levels with protein expression across tissues, identifying potential tissue-specific post-transcriptional regulatory mechanisms.

  • Metabolomic Integration: Include tissue-specific metabolomic profiling to connect TP53I3 function with metabolic phenotypes across different tissues.

Advanced Protein Interaction Studies:

  • Tissue-Specific Interactome Analysis: Employ BioID, APEX, or other proximity labeling techniques to identify tissue-specific protein interaction partners of TP53I3, revealing context-dependent functional networks.

  • Domain-Specific Interaction Mapping: Develop approaches to map interactions of specific TP53I3 domains with other proteins across tissues, providing insights into structural determinants of tissue-specific functions.

  • In Situ Protein Interaction Visualization: Advance techniques like proximity ligation assay for visualizing TP53I3 protein interactions within intact tissue specimens.

Translational Research Approaches:

  • Tissue-Specific Biomarker Development: Create a panel of tissue-specific TP53I3-related biomarkers that account for context-dependent functions in different organs.

  • Patient-Derived Models: Establish libraries of patient-derived xenografts or organoids from different tissue origins with characterized TP53I3 status to evaluate tissue-specific therapeutic responses.

  • Clinical Sample Analysis: Develop multiplex imaging approaches to simultaneously assess TP53I3 expression, p53 status, and tissue-specific markers in patient samples.

These methodological advances would collectively provide a more comprehensive understanding of how TP53I3 functions across different tissues, potentially explaining why its role varies between cancer types—from tumor suppression in some contexts to potential oncogenic effects in others . This tissue-specific perspective is essential for developing precisely targeted therapeutic approaches that account for contextual differences in TP53I3 biology.

How do genetic and epigenetic variations in TP53I3 contribute to cancer risk across different populations?

Understanding how genetic and epigenetic variations in TP53I3 contribute to cancer risk across populations requires a comprehensive approach that integrates population genetics, molecular epidemiology, and functional genomics. This complex relationship remains incompletely characterized, but several methodological strategies could advance our understanding:

Population-Scale Genetic Analysis:

  • Genome-Wide Association Studies (GWAS): Conduct large-scale GWAS focused on TP53I3 variants across ethnically diverse populations to identify risk-associated polymorphisms. While germline variants in TP53I3 associated with cancer risk are still being investigated , systematic population studies could reveal previously unrecognized associations.

  • Whole Genome/Exome Sequencing: Perform deep sequencing of TP53I3 in case-control cohorts from diverse populations to identify rare variants that might be missed by GWAS approaches but could have substantial functional effects.

  • Population-Specific Variant Cataloging: Develop comprehensive catalogs of TP53I3 variants specific to different ethnic populations, including frequency data and predicted functional impacts.

Functional Characterization of Population Variants:

  • High-Throughput Functional Screening: Develop multiplexed assays to simultaneously test the functional impact of multiple TP53I3 variants on DNA repair efficiency, ROS regulation, and drug sensitivity.

  • Structure-Function Relationship Analysis: Map population variants onto the protein structure of TP53I3 to predict how specific variants might affect protein function based on their location in functional domains.

  • Isogenic Cell Line Models: Create panels of isogenic cell lines harboring population-specific TP53I3 variants to directly compare their functional effects in a controlled genetic background.

Epigenetic Variation Analysis:

  • Population Epigenomics: Characterize epigenetic patterns (DNA methylation, histone modifications) at the TP53I3 locus across different populations to identify epigenetic variations that might influence gene expression.

  • Environmental Interaction Studies: Investigate how environmental factors relevant to specific populations interact with TP53I3 epigenetic regulation, potentially explaining population-specific cancer risk profiles.

  • Longitudinal Epigenetic Monitoring: Conduct studies that track changes in TP53I3 epigenetic marks over time in different populations, correlating these changes with cancer incidence.

Integrative Risk Assessment:

  • Polygenic Risk Scores: Develop population-specific polygenic risk scores that incorporate TP53I3 variants along with other cancer risk loci to provide more accurate personalized risk assessment.

  • Gene-Environment Interaction Modeling: Create mathematical models that account for interactions between TP53I3 genetic/epigenetic variations and population-specific environmental exposures.

  • Multi-Ancestry Meta-Analysis: Perform meta-analyses across studies from different ancestral populations to identify both shared and population-specific TP53I3 risk variants.

Translational Applications:

  • Population-Adapted Screening Protocols: Develop cancer screening guidelines that account for population-specific TP53I3 variation patterns and associated risk profiles.

  • Precision Prevention Strategies: Design preventive interventions tailored to individuals with specific TP53I3 risk variants, particularly in high-risk populations.

  • Pharmacogenomic Applications: Investigate how population-specific TP53I3 variants affect response to cancer preventive agents or treatments.

Research Considerations and Challenges:
The current literature indicates that TP53I3 may have diverse roles across different cancer types, with overexpression correlating with increased survival in breast cancer while loss of expression promotes other cancers like NSCLC, colon, and papillary thyroid cancer . This cancer-type specificity may intersect with population differences in cancer susceptibility, requiring careful interpretation of genetic associations.

Additionally, the functional consequences of TP53I3 variants appear to be complex. For example, the p.S252X variant affects homologous recombination repair efficiency and sensitivity to chemotherapeutic drugs , but the prevalence and impact of this variant across different populations remain to be fully characterized.

Future research should prioritize inclusive sampling across diverse populations, functional validation of population-specific variants, and integration of genetic and epigenetic data to comprehensively assess how TP53I3 variations contribute to differential cancer risk across human populations.

Product Science Overview

Function and Mechanism

TP53I3 is similar to oxidoreductases, enzymes that play a role in cellular responses to oxidative stress and irradiation . The protein is involved in p53-mediated cell death, which is a critical mechanism for preventing the proliferation of cells with damaged DNA . When cells are exposed to genotoxic agents, TP53I3 is localized to the cytoplasm and is induced in both primary and transformed cell cultures .

Role in Cancer

The TP53 gene, which encodes the p53 protein, is mutated in approximately half of all human cancers . Mutations in this gene often lead to the loss of p53’s tumor-suppressing functions, resulting in uncontrolled cell proliferation and tumor development . TP53I3, being a downstream target of p53, is also implicated in these processes. The protein’s role in oxidative stress response and apoptosis makes it a significant player in the cellular mechanisms that prevent cancer progression .

Research and Therapeutic Potential

Given its involvement in p53-mediated pathways, TP53I3 is a subject of interest in cancer research. Understanding its function and regulation could lead to new therapeutic strategies aimed at restoring p53’s tumor-suppressing activities in cancers where TP53 is mutated . Additionally, TP53I3’s role in oxidative stress response suggests potential applications in therapies targeting cellular redox balance .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.