The GSTP1 antibody is a research and diagnostic tool designed to detect glutathione S-transferase pi 1 (GSTP1), a phase II detoxification enzyme encoded by the GSTP1 gene. GSTP1 plays critical roles in cellular defense mechanisms, including conjugation of glutathione to electrophilic compounds, regulation of oxidative stress, and modulation of signaling pathways (e.g., Akt, MAPK) . The antibody is used in immunological assays to study GSTP1 expression, subcellular localization, and clinical correlations in cancer, drug resistance, and therapeutic response .
CSC Regulation: GSTP1 sustains cancer stem cell (CSC) phenotypes via CaMK2A/NRF2 signaling, promoting self-renewal and metastasis .
Oxidative Stress: GSTP1 reduces reactive oxygen species (ROS), enabling tumor adaptation to hypoxia and radiation .
GSTP1 antibodies enable precise monitoring of GSTP1 in clinical trials targeting CSCs or drug-resistant tumors. For example:
GSTP1, a polymorphic gene, encodes active GSTP1 variant proteins with functional differences. These proteins are believed to play a role in xenobiotic metabolism and may contribute to susceptibility to cancer and other diseases. As a glutathione S-transferase belonging to the pi class, GSTP1 is part of the GST family, whose enzymes are crucial for detoxification. They catalyze the conjugation of various hydrophobic and electrophilic compounds with reduced glutathione. Soluble GSTs are classified into four main classes based on their biochemical, immunological, and structural properties: alpha, mu, pi, and theta. GSTP1 catalyzes the reaction between glutathione and an acceptor molecule, forming Sulfur-substituted glutathione. Reactions involving glutathione contribute to transforming a wide range of electrophiles, such as reactive products of lipids, proteins, carcinogens, therapeutic drugs, environmental toxins, and oxidative stress products. Frequent inactivation of GSTP1 due to CpG hypermethylation in pituitary adenomas suggests a potential role in aggressive tumor behavior. Notably, GSTP1 may be a transcriptional target of the tumor suppressor gene p53. Single-nucleotide polymorphisms in GSTP1 are linked to altered protein binding, influencing its role in carcinogen and drug metabolism, potentially impacting disease pathogenesis and drug response. GST-pi might play a central role in the proliferation of androgen-independent prostate cancer cells.
The solution contains 1mg/ml of GSTP1 antibody in a buffer consisting of PBS at pH 7.4, 10% Glycerol, and 0.02% Sodium Azide.
The GSTP1 antibody has been validated for use in various applications, including ELISA, Western blot analysis, ICC/IF, and Flow cytometry. These tests confirm its specificity and reactivity. However, it is important to note that optimal antibody dilutions should be determined empirically for each specific application to achieve the best results.
Glutathione S-transferase P 1, Gst P1, GST YF-YF, GST class-pi, GST-piB, Preadipocyte growth factor.
GSTP1 antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
PAT12C10AT
Anti-human GSTP1 mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human GSTP1 amino acids 1-210 purified from E. coli.
Mouse IgG1 heavy chain and κ light chain.
GSTP1 is a member of the Glutathione S-transferase family that plays a crucial role in the metabolic detoxification of various environmental carcinogens and lipid hydroperoxides. This enzyme is essential for cellular protection against oxidative stress by neutralizing reactive oxygen species and harmful electrophilic compounds. GSTP1 becomes upregulated in response to oxidative stress, highlighting its importance in maintaining cellular homeostasis .
Additionally, the GSTP1 gene is subject to CpG island hypermethylation, a modification that has been associated with human prostatic carcinogenesis and other cancer types . This epigenetic modification can be detected in biological fluids such as urine, ejaculate, and plasma from men diagnosed with prostate cancer, making GSTP1 a valuable biomarker for cancer research .
Based on current research resources, several types of GSTP1 antibodies are available for research purposes:
Monoclonal antibodies:
GSTP1 Antibody (F-6): A mouse monoclonal IgG2a antibody that detects human GSTP1 through Western blotting (WB), immunoprecipitation (IP), immunofluorescence (IF), and enzyme-linked immunosorbent assay (ELISA)
GSTP1 Antibody (3F2C2): A mouse monoclonal IgG1 kappa light chain antibody validated for WB, IP, IF, and immunohistochemistry with paraffin-embedded sections (IHC-P)
Various conjugated forms:
The key differences between these antibodies include their isotype (IgG1 vs. IgG2a), epitope recognition, and validated applications. Selection should be based on the intended experimental methodology, target species, and specific research objectives.
Before implementing GSTP1 antibodies in a new research system, several validation steps are essential:
Specificity verification:
Test antibodies on positive control samples (cells/tissues known to express GSTP1)
Include negative controls (samples with known GSTP1 silencing via promoter hypermethylation)
Verify the expected molecular weight (~23 kDa) in Western blotting applications
Cross-validation approaches:
Application-specific validation:
For Western blotting: Optimize sample preparation conditions (reducing vs. non-reducing), antibody dilutions, and detection methods
For IHC/IF: Determine optimal antigen retrieval methods, antibody concentrations, and incubation times
For cancer tissue analysis: Be aware that GSTP1 silencing through promoter hypermethylation is common in certain cancers (90-95% of prostate cancers)
Thorough validation ensures reliable and reproducible results when implementing GSTP1 antibodies in new experimental systems.
For optimal Western blot detection of GSTP1, researchers should follow this methodological approach:
Sample preparation:
Electrophoresis and transfer:
Antibody incubation:
Controls and interpretation:
This protocol has been validated in multiple research contexts and optimizes detection sensitivity while minimizing background interference.
Methylation-specific PCR (MSP) is the established technique for assessing GSTP1 promoter methylation. Based on published methodologies, researchers should follow this approach:
DNA extraction and bisulfite modification:
PCR amplification:
Analysis:
Validation:
This methodology allows for reliable assessment of GSTP1 methylation status, which is particularly relevant in cancer research where GSTP1 promoter hypermethylation is a frequent event.
For effective immunohistochemical detection of GSTP1, researchers should optimize these critical parameters:
Tissue preparation:
Use formalin-fixed, paraffin-embedded (FFPE) sections at 4-6 μm thickness
Implement standardized fixation protocols to ensure consistent epitope preservation
Antigen retrieval:
Perform heat-induced epitope retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Optimization of retrieval conditions may be necessary depending on fixation time and tissue type
Antibody selection and incubation:
Use antibodies validated for IHC-P, such as the GSTP1 Antibody (3F2C2)
Optimize antibody dilution through titration experiments
Include appropriate controls:
Positive control tissues (normal tissues known to express GSTP1)
Negative controls (tissues with known GSTP1 silencing or omission of primary antibody)
Detection system:
Select an appropriate detection system (e.g., polymer-based systems for increased sensitivity)
Develop signal using DAB or other suitable chromogen
Counterstain with hematoxylin for nuclear visualization
Interpretation considerations:
Evaluate cytoplasmic and potentially nuclear staining patterns
Quantify staining intensity and percentage of positive cells
Be aware that GSTP1 expression may be lost due to promoter hypermethylation in certain cancers
Validation with mRNA in situ hybridization may be valuable as demonstrated in research showing concordance between GSTP1 mRNA expression and protein staining
Attention to these parameters ensures reliable and reproducible immunohistochemical detection of GSTP1 in tissue samples.
GSTP1 expression exhibits significant variations across different cancer types through several key mechanisms:
Epigenetic silencing:
Race-associated expression patterns:
GSTP1-positive prostate cancers are significantly more common in Black men compared to White men
Research shows that 14.9% of prostate adenocarcinomas from Black patients had GSTP1-positive tumor microarray cores, which was 2.5 times higher than the percentage from White patients (5.9%)
The percentage of tumors from Black patients with all tumor microarray spots positive for GSTP1 (9.5%) was 3-fold higher than the percentage from White patients (3.2%)
Association with molecular subtypes:
These variations in GSTP1 expression patterns have significant implications for understanding cancer biology and developing targeted therapeutic approaches, particularly in addressing cancer disparities among different populations.
GSTP1 phosphorylation represents a critical intersection between detoxification processes and cancer signaling pathways:
EGFR-mediated GSTP1 phosphorylation:
Experimental protocols for studying GSTP1 phosphorylation:
Cell-free systems using recombinant GSTP1, EGFR active kinase domain, and ATP can demonstrate this phosphorylation
Protocols typically involve preincubating GSTP1 with GSH (5 mM) to mimic intracellular conditions
Analysis requires non-reducing SDS-PAGE conditions followed by Western blotting with anti-phosphotyrosine antibodies
Connection to JNK signaling pathway:
Implications for cancer biology:
GSTP1 has been described as a "caretaker gene whose loss increases cell survival in response to protracted oxidative stress"
The phosphorylation state of GSTP1 may influence its ability to regulate cellular stress responses and survival pathways
This positions GSTP1 at the intersection of detoxification processes and signaling pathways that control cell fate
This connection between GSTP1 phosphorylation and signaling pathways suggests more complex roles for GSTP1 in cancer biology beyond its classical detoxification function.
GSTP1 polymorphisms significantly impact enzyme function and cancer susceptibility through several mechanisms:
These findings highlight the complex relationship between GSTP1 genetic variations and cancer susceptibility, involving both direct effects on enzyme activity and potential interactions with environmental and epigenetic factors.
Designing robust experiments to investigate GSTP1's role in oxidative stress response requires multifaceted approaches:
Cellular models and manipulation strategies:
Generate cellular models with varied GSTP1 expression levels:
Oxidative stress induction protocols:
Implement controlled oxidative stress conditions using:
H₂O₂ treatment at various concentrations and time points
Glutathione depletion using buthionine sulfoximine
Exposure to environmental carcinogens or lipid hydroperoxides
Measure cellular responses using:
Cell viability assays (MTT, XTT)
ROS detection (DCF-DA fluorescence, EPR spectroscopy)
Lipid peroxidation markers (MDA, 4-HNE levels)
Signaling pathway analysis:
Investigate GSTP1's interaction with JNK pathway:
Examine EGFR-GSTP1 interaction:
EGFR-mediated GSTP1 phosphorylation assays
Effects of EGFR inhibitors on GSTP1 function
Impact of GSTP1 phosphorylation on its detoxification activity
Integrated multi-omics approaches:
Transcriptomic analysis to identify GSTP1-dependent gene expression changes under oxidative stress
Proteomic studies to detect GSTP1 interaction partners in stress response pathways
Metabolomic analysis focusing on glutathione metabolism and detoxification products
These comprehensive experimental approaches will enable researchers to elucidate GSTP1's multifaceted roles in oxidative stress responses beyond its classical detoxification function.
To identify novel GSTP1 post-translational modifications (PTMs), researchers should implement these advanced methodological approaches:
Mass spectrometry-based PTM profiling:
Immunoprecipitate GSTP1 using validated antibodies such as GSTP1 Antibody (F-6) or (3F2C2)
Perform tryptic digestion followed by LC-MS/MS analysis
Implement database searches with variable modifications including phosphorylation, acetylation, ubiquitination, and SUMOylation
Use targeted MS approaches (PRM/MRM) for quantitative analysis of specific modifications
Phosphorylation-specific analytical techniques:
Expand on established EGFR-mediated GSTP1 phosphorylation protocols :
Use recombinant GSTP1 preincubated with 5 mM GSH
Incubate with various kinases (beyond EGFR) in kinase buffer with ATP
Analyze by non-reducing SDS-PAGE and Western blotting with phospho-specific antibodies
Implement phospho-proteomics approaches:
Phospho-enrichment techniques (IMAC, TiO₂)
Phospho-specific antibodies against common motifs
Stress-induced modification analysis:
Expose cells to various stressors (oxidative stress, electrophiles, chemotherapeutics)
Compare GSTP1 modification profiles before and after stress induction
Correlate modifications with changes in GSTP1 activity, localization, or protein interactions
Site-directed mutagenesis validation:
Generate GSTP1 mutants at putative modification sites
Express wild-type and mutant forms in cellular models
Compare functional consequences of mutations on:
Catalytic activity
Protein stability
Protein-protein interactions (particularly with JNK and EGFR)
Cellular stress responses
These methodological approaches will enable identification of novel GSTP1 PTMs and their functional significance in cellular physiology and pathological conditions, potentially revealing new therapeutic targets and biomarkers.
To examine GSTP1's dual role in detoxification and cell signaling, researchers should implement these sophisticated experimental approaches:
Protein interaction network analysis:
Perform comprehensive immunoprecipitation studies:
Map interactions under various conditions:
Basal vs. stress conditions
Wild-type vs. mutant GSTP1
Phosphorylated vs. non-phosphorylated GSTP1
Functional investigation of GSTP1-JNK interaction:
Implement cell-free JNK kinase assays as described in the literature:
Extend to cellular systems:
Compare JNK pathway activation in GSTP1-positive vs. GSTP1-negative cells
Assess impact of GSTP1 phosphorylation state on JNK inhibition
Examine consequences for apoptosis and stress responses
GSTP1 role in EGFR signaling modulation:
Investigate bidirectional relationship between GSTP1 and EGFR:
EGFR-mediated GSTP1 phosphorylation
Potential GSTP1 effects on EGFR signaling outputs
Examine functional consequences in relevant cancer models:
Integration of detoxification and signaling functions:
Design experiments to test how GSTP1's detoxification capacity affects its signaling roles:
Enzyme activity assays with various substrates
GSH depletion/repletion studies
Structure-function analyses using GSTP1 mutants with altered catalytic properties
Examine how signaling events modify GSTP1's detoxification function:
Effect of phosphorylation on enzyme kinetics
Changes in substrate specificity
Alterations in protein stability or cellular localization
These integrated approaches will help elucidate GSTP1's complex functions at the intersection of detoxification processes and signaling pathways, potentially revealing novel therapeutic strategies targeting GSTP1 in various disease contexts.
Research has revealed significant racial differences in GSTP1 expression patterns, particularly in prostate cancer. When investigating these differences, researchers should consider several critical methodological factors:
Sample collection and representation:
Ensure adequate representation of diverse racial and ethnic groups
Published research found that 14.9% of prostate adenocarcinomas from Black patients had GSTP1-positive TMA cores compared to only 5.9% from White patients
Sample sizes must be sufficient to detect statistically significant differences (the cited study examined 1673 primary prostatic adenocarcinomas)
Multidimensional analysis approach:
Control for confounding variables:
Account for potentially confounding factors:
Age, socioeconomic status, and access to healthcare
Tumor grade, stage, and molecular subtypes
Treatment history and comorbidities
Statistical approaches should include multivariate analyses
Molecular context consideration:
Biological and clinical implications:
Connect molecular findings to biological hypotheses
Published research suggested that GSTP1-positive cases may "represent a distinct molecular subtype of prostate cancer" that could provide "a biological underpinning for the observed disparate outcomes for Black men"
Explore therapeutic and prognostic implications of observed differences
These methodological considerations ensure robust and clinically relevant investigations of racial differences in GSTP1 expression patterns, potentially contributing to addressing cancer disparities.
When encountering inconsistent GSTP1 detection in cancer samples, researchers should implement these troubleshooting approaches:
Address biological variability factors:
Heterogeneous GSTP1 promoter methylation:
Intratumoral heterogeneity:
Optimize technical parameters:
Antibody selection and validation:
Antigen retrieval optimization:
Test multiple antigen retrieval methods (citrate vs. EDTA buffers)
Optimize heating time and temperature
Detection system sensitivity:
Implement signal amplification methods for low-expressing samples
Consider more sensitive detection systems (polymer-based vs. ABC method)
Implement multi-modal confirmation:
Correlate protein with mRNA expression:
Integrate with methylation analysis:
Correlate protein expression with promoter methylation status
Understand that hypermethylation is often associated with loss of expression
Controls and standardization:
Include consistent positive and negative controls in each experiment
Implement standardized scoring systems for immunohistochemistry
Document all technical parameters to ensure reproducibility
These troubleshooting approaches address both biological and technical factors contributing to inconsistent GSTP1 detection, leading to more reliable and interpretable results in cancer research.
Integrating GSTP1 genotype, expression, and methylation data requires robust methodological approaches and analytical frameworks:
Unified sample processing strategy:
Implement DNA/RNA/protein co-extraction protocols from the same specimen
Document all preservation methods and extraction conditions
Maintain consistent sample identifiers across all analyses
Standardized methodological approaches:
Genotyping: Use PCR-RFLP for Ile105Val polymorphism detection
Methylation analysis: Implement methylation-specific PCR
Expression analysis:
Data integration framework:
Implement consistent quantification methods:
For IHC: Standardized scoring of staining intensity and percentage
For methylation: Binary (methylated/unmethylated) or quantitative (% methylation)
For genotyping: Categorize as homozygous wild-type, heterozygous, or homozygous variant
Statistical integration approaches:
Correlation analyses between methylation status and expression
Association tests between genotype and methylation/expression
Multivariate models incorporating all molecular data with clinical parameters
Validation strategies:
Internal validation through technical replicates
Cross-platform validation
Verify methylation effects on expression using multiple techniques
Confirm genotype-phenotype associations across different experimental systems
Data interpretation considerations:
Consider context-dependent effects:
Account for potential independence of mechanisms:
These integrated approaches enable comprehensive characterization of GSTP1 alterations across genetic, epigenetic, and protein levels, providing deeper insights into GSTP1 biology in health and disease contexts.
Glutathione S-Transferase pi 1 (GSTP1) is a member of the glutathione S-transferase (GST) family, which plays a crucial role in the detoxification of endogenous and exogenous compounds. GSTP1 is involved in the conjugation of glutathione to various electrophilic compounds, including metabolites generated by oxidative processes, environmental toxins, carcinogens, and anti-cancer drugs .
GSTP1 is highly polymorphic and encodes functionally different variant proteins that are thought to be involved in xenobiotic metabolism and susceptibility to cancer and other diseases . In humans, GSTP1 expression is found in various tissues, including the liver, where it is primarily expressed in non-parenchymal cells such as bile duct cells, Kupffer cells, macrophages, and endothelial cells . In contrast, mouse GSTP1 (mGstp) is highly expressed in hepatocytes .
The Mouse Anti-Human GSTP1 antibody is a monoclonal antibody that specifically detects human GSTP1 in various applications, including Western blotting, immunocytochemistry, and ELISA . This antibody is produced by immunizing mice with recombinant human GSTP1 protein and is purified using protein A or G from hybridoma culture supernatant .
The Mouse Anti-Human GSTP1 antibody is widely used in research to study the expression and function of GSTP1 in different tissues and under various conditions. For example, it has been used to detect GSTP1 in human cervical epithelial carcinoma cell lines (HeLa) and human embryonic stem cells differentiated into hepatocytes . Additionally, this antibody has been employed in studies investigating the role of GSTP1 in liver toxicity and drug metabolism .