GSTA4 Human

Glutathione S-Transferase Alpha 4 Human Recombinant
Shipped with Ice Packs
In Stock

Description

GSTA4 Human Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 246 amino acids (1-222) and having a molecular mass of 28.3kDa.
GSTA4 is fused to a 24 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Glutathione S-transferase A4 (GSTA4), a member of the GST superfamily, plays a crucial role in cellular defense mechanisms. It detoxifies various harmful compounds, including toxins, carcinogens, and reactive electrophiles. GSTA4 exhibits high activity towards reactive carbonyl compounds like alk-2-enals and efficiently catalyzes the conjugation of reduced glutathione to 4-hydroxynonenal, a biomarker for oxidative stress. Its expression is particularly high in the brain, placenta, and skeletal muscle, while being lower in the lungs and liver.
Description
Recombinant human GSTA4, expressed in E.coli, is a single, non-glycosylated polypeptide chain with 246 amino acids (1-222) and a molecular weight of 28.3 kDa. The protein is purified using proprietary chromatographic techniques and is fused to a 24 amino acid His-tag at the N-terminus.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The GSTA4 solution is provided at a concentration of 1 mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0), 20% glycerol, 2mM DTT, and 100mM NaCl.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended periods, storage at -20°C is recommended. The addition of a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of GSTA4 is greater than 95.0%, as determined by SDS-PAGE analysis.
Synonyms
Glutathione S-transferase A4, GST class-alpha member 4, Glutathione S-transferase A4-4, GSTA4, GSTA4-4.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMAARPK LHYPNGRGRM ESVRWVLAAA GVEFDEEFLE TKEQLYKLQD GNHLLFQQVP MVEIDGMKLV QTRSILHYIA DKHNLFGKNL KERTLIDMYV EGTLDLLELL IMHPFLKPDD QQKEVVNMAQ KAIIRYFPVF EKILRGHGQS FLVGNQLSLA
DVILLQTILA LEEKIPNILS AFPFLQEYTV KLSNIPTIKR FLEPGSKKKP PPDEIYVRTV YNIFRP.

Q&A

What is GSTA4 and what is its primary function in human cells?

GSTA4 (Glutathione S-Transferase Alpha 4) is a phase II detoxifying enzyme belonging to the alpha class of glutathione S-transferases. It plays a crucial role in cellular detoxification processes by catalyzing the conjugation of glutathione to various xenobiotics and endogenous compounds. GSTA4 specifically excels at detoxifying 4-hydroxynonenal (4-HNE), a major byproduct of lipid peroxidation and marker of oxidative stress . This enzyme represents one of five isozymes in the human alpha class GST family, with each member responsible for detoxifying different compounds including carcinogens, mutagens, chemotherapeutic drugs, steroids, and various byproducts of oxidative stress . The catalytic efficiency of GSTA4 toward 4-HNE is significantly higher than other GST enzymes, making it a critical player in cellular defense against oxidative damage.

How is GSTA4 expression regulated in normal and pathological conditions?

GSTA4 expression is tightly regulated through multiple mechanisms, with significant differences between normal and pathological states. In colorectal cancer, GSTA4 is overexpressed in both adenomas and invasive carcinomas compared to normal tissue . This overexpression is regulated by the oncogenic transcription factor activator protein 1 (AP-1), which becomes activated during malignant transformation . Under normal physiological conditions, GSTA4 expression typically responds to oxidative stress through activation of Nrf2 (Nuclear factor erythroid 2-related factor 2) pathway, which serves as a master regulator of cellular responses to oxidative insults. In pathological contexts such as cancer, aberrant signaling pathways including NF-κB and MAPK cascades can further influence GSTA4 expression. Researchers investigating GSTA4 regulation should consider employing promoter analysis, ChIP assays, and reporter gene assays to fully characterize the transcriptional mechanisms controlling GSTA4 expression in their specific tissue or disease model of interest.

What are the established experimental models for studying GSTA4 function?

Several well-established experimental models exist for investigating GSTA4 function. In vitro models include human cancer cell lines such as HCT116 colorectal cancer cells, which have been successfully manipulated using CRISPR/Cas9 gene editing to create GSTA4-deficient variants (HCT116 ΔGSTA4) . Such gene editing approaches typically target exons 1 and 2 of the GSTA4 gene to create homozygous deletions . Primary cell cultures transfected with GSTA4-expressing plasmids provide complementary systems to study gain-of-function effects. For in vivo studies, xenograft models using BALB/cJGpt-Foxn1 nu/Gpt nude mice have demonstrated decreased tumor growth with GSTA4-deficient cancer cells . Additionally, GSTA4 knockout mouse models are valuable for studying systemic effects of GSTA4 deficiency, particularly in skin cancer and other malignancies. When designing GSTA4 studies, researchers should carefully select appropriate models based on their specific research questions, considering species differences in GSTA4 expression patterns and the limitations of each model system in recapitulating human disease states.

How does GSTA4 mechanistically influence cancer cell proliferation and chemoresistance?

GSTA4 influences cancer cell proliferation and chemoresistance through multiple interconnected molecular mechanisms. Research using GSTA4-inactivated HCT116 colorectal cancer cells has revealed that GSTA4 activates key proliferative signaling pathways, particularly PI3K/AKT and p38 MAPK . When GSTA4 is inactivated in these cells, phosphorylation of both AKT and p38 MAPK significantly decreases, suggesting that GSTA4 normally promotes continuous activation of these pathways . This mechanistic relationship appears to be independent of GSTA4's detoxification function, as the slight increases in reactive oxygen species (ROS) observed in GSTA4-deficient cells did not correlate with proliferation changes .

In terms of chemoresistance, GSTA4 inactivation increases cancer cell susceptibility to both 5-fluorouracil (5-FU) and oxaliplatin, though through different mechanisms . With 5-FU treatment, GSTA4-deficient cells show significantly increased γH2AX (a marker of double-stranded DNA breaks), indicating enhanced DNA damage . In contrast, oxaliplatin-induced DNA crosslinking shows less dramatic differences between GSTA4-deficient and normal cells . These findings suggest that GSTA4 might confer chemoresistance not only through its detoxification activity but also through modulation of DNA damage response pathways.

Researchers investigating these mechanisms should consider using phosphoproteomic approaches to comprehensively map GSTA4-dependent signaling networks, protein-protein interaction studies to identify direct binding partners of GSTA4, and high-resolution DNA damage assays to clarify the relationship between GSTA4 and genotoxic stress response.

What methodologies are most effective for genetic manipulation of GSTA4 in experimental systems?

The most effective methodologies for genetic manipulation of GSTA4 depend on the specific research objectives and experimental system. For knockout studies in cell lines, CRISPR/Cas9 gene editing has proven highly effective. The approach detailed in recent research utilized the lentiCRISPRv2 system with guide RNAs (gRNAs) targeting exons 1 and 2 of the GSTA4 gene . Specifically, co-transfection with gRNA66 and gRNA214 successfully created a 1,135 bp homozygous deletion across these exons . The procedure involves:

  • Design of gRNAs using specialized tools like CRISPRdirect (https://crispr.dbcls.jp)

  • Cloning gRNAs into the pLentiCRISPRv2 vector

  • Co-transfection of target cells using Lipofectamine 3000 reagent

  • Selection of transfected cells with puromycin (4 μg/ml initially, then 1 μg/ml for maintenance)

  • Screening of clones via PCR amplification and Sanger sequencing

For transient knockdown experiments, siRNA or shRNA approaches provide alternatives with less potential for off-target effects. For overexpression studies, transfection with GSTA4-expressing plasmids under various promoters allows investigation of dose-dependent effects.

In in vivo models, conditional knockout systems using Cre-loxP technology offer tissue-specific and temporally controlled GSTA4 manipulation. Adeno-associated virus (AAV) vectors can provide localized GSTA4 modification in specific tissues or tumors.

Validation of successful genetic manipulation should include both genomic PCR confirmation and functional assays such as 4-HNE sensitivity testing, as 4-HNE is a specific substrate for GSTA4 .

How do GSTA4 polymorphisms influence cancer susceptibility in human populations?

GSTA4 polymorphisms significantly influence cancer susceptibility across human populations, with particular relevance to skin cancer. Single-nucleotide polymorphisms (SNPs) in the GSTA4 gene have been associated with increased risk of nonmelanoma skin cancer (NMSC) . The tag SNP-based approach has proven effective in identifying these cancer-associated variants .

Research indicates that GSTA4 is a novel susceptibility gene for NMSC, affecting risk in both mice and humans . This dual-species evidence strengthens the biological plausibility of GSTA4's role in cancer development. Polymorphisms that reduce GSTA4 activity or expression likely increase cancer risk by diminishing cellular capacity to detoxify 4-HNE and other harmful byproducts of oxidative stress, leading to elevated levels of DNA damage and protein modification.

For researchers investigating GSTA4 polymorphisms, methodologically sound approaches include:

  • Case-control studies with careful matching for relevant demographic and environmental factors

  • Tag SNP selection to efficiently capture genetic variation across the GSTA4 locus

  • Functional characterization of identified variants using reporter assays, protein activity measurements, and structural modeling

  • Integration with gene-environment interaction analyses, particularly focusing on UV exposure for skin cancer studies

  • Multi-ethnic cohort studies to assess population-specific effects of GSTA4 variants

These approaches can help distinguish causative variants from those in linkage disequilibrium and provide mechanistic insights into how specific polymorphisms alter GSTA4 function or expression.

What is the relationship between GSTA4 and reactive oxygen species (ROS) in cancer progression?

The relationship between GSTA4 and reactive oxygen species (ROS) in cancer progression is complex and context-dependent. GSTA4 primarily functions as a detoxifying enzyme that neutralizes 4-hydroxynonenal (4-HNE), a major byproduct of lipid peroxidation resulting from oxidative stress . Studies in colorectal cancer cells have demonstrated that inactivation of GSTA4 increases baseline ROS production compared to GSTA4-expressing cells . This elevation in ROS is further amplified when GSTA4-deficient cells are treated with chemotherapeutic agents like 5-FU and oxaliplatin .

Interestingly, the relationship between GSTA4, ROS, and cell proliferation appears to be distinct from the classic model where increased ROS drives proliferation. In colorectal cancer studies, despite increased ROS in GSTA4-deficient cells, these cells exhibited reduced proliferation, suggesting that GSTA4's pro-proliferative effects operate primarily through direct regulation of signaling pathways rather than through ROS modulation .

For researchers studying this relationship, methodological considerations should include:

  • Using multiple ROS detection methods to overcome limitations of individual assays

  • Employing antioxidant rescue experiments to establish causality between ROS and observed phenotypes

  • Measuring specific ROS species rather than general oxidative stress

  • Considering subcellular localization of ROS production and GSTA4 activity

  • Integrating transcriptomic and metabolomic approaches to comprehensively profile oxidative stress responses

What are the potential therapeutic implications of targeting GSTA4 in cancer treatment?

Targeting GSTA4 represents a promising therapeutic approach in cancer treatment, particularly for colorectal cancer where GSTA4 is overexpressed and contributes to both proliferation and chemoresistance . Several lines of evidence support GSTA4 as a therapeutic target:

  • Chemosensitization Effect: GSTA4 inactivation increases cancer cell susceptibility to standard chemotherapeutic agents, including 5-fluorouracil (5-FU) and oxaliplatin . This suggests that GSTA4 inhibitors could serve as chemosensitizing agents, allowing for lower doses of conventional chemotherapeutics and potentially reducing their side effects.

  • Anti-proliferative Activity: GSTA4 deficiency reduces cancer cell proliferation by blocking the activation of AKT and p38 MAPK pathways . This anti-proliferative effect was confirmed in both in vitro studies and xenograft models, where GSTA4-deficient tumors showed reduced growth compared to GSTA4-expressing tumors .

  • Tumor-Specific Targeting Potential: The overexpression of GSTA4 in tumor tissues compared to normal tissues provides an opportunity for selective targeting of cancer cells, potentially minimizing effects on normal tissues.

For researchers exploring GSTA4-targeted therapies, several methodological approaches should be considered:

  • Development of small molecule inhibitors specific to GSTA4, distinguishing it from other GST family members

  • Exploration of targeted degradation approaches such as PROTACs (Proteolysis Targeting Chimeras)

  • Investigation of antisense oligonucleotides or siRNA delivery systems for GSTA4 knockdown

  • Combination therapy studies testing GSTA4 inhibition with conventional chemotherapeutics

  • Biomarker development to identify patients most likely to benefit from GSTA4-targeted therapy

The table below summarizes key experimental findings relevant to GSTA4 as a therapeutic target:

Experimental ApproachKey FindingTherapeutic Implication
GSTA4 knockout in HCT116 cellsDecreased cell proliferation and increased ROSGSTA4 inhibition may have direct anti-tumor effects
Chemotherapy treatment of GSTA4-deficient cellsIncreased susceptibility to 5-FU and oxaliplatinGSTA4 inhibition may enhance chemotherapy efficacy
Xenograft model with GSTA4-deficient cellsReduced tumor growth in vivoGSTA4 targeting may be effective in intact organisms
Signaling pathway analysisBlocked AKT and p38 MAPK activationGSTA4 inhibition affects multiple cancer-promoting pathways
4-HNE sensitivity testingIncreased 4-HNE-induced cell killing in GSTA4-deficient cellsGSTA4 inhibition may synergize with therapies inducing oxidative stress

What are the most reliable approaches for measuring GSTA4 activity in biological samples?

Measuring GSTA4 activity in biological samples requires careful consideration of both general GST activity and GSTA4-specific activity. Since GSTA4 specifically excels at conjugating glutathione to 4-hydroxynonenal (4-HNE), assays utilizing 4-HNE as a substrate provide the most reliable measure of GSTA4-specific activity . Several methodological approaches can be employed:

  • Spectrophotometric Assays: These measure the conjugation of glutathione to 4-HNE by monitoring absorbance changes. While accessible, these assays may lack specificity when other GSTs have overlapping substrate preferences.

  • High-Performance Liquid Chromatography (HPLC): This approach separates and quantifies glutathione-4-HNE conjugates, offering improved specificity over spectrophotometric methods.

  • Cell Viability Assays with 4-HNE: As demonstrated in research, cells deficient in GSTA4 show increased sensitivity to 4-HNE-induced cell death . Comparative cell viability assays following 4-HNE treatment can serve as functional indicators of GSTA4 activity.

  • Mass Spectrometry: LC-MS/MS approaches provide the highest specificity for measuring glutathione-4-HNE conjugates and can detect even small changes in GSTA4 activity.

  • Immunoprecipitation Followed by Activity Assay: This approach combines antibody-based isolation of GSTA4 protein with subsequent activity measurement, helping to distinguish GSTA4 activity from other GSTs.

For reliable measurements, researchers should consider several factors:

  • Sample preparation techniques that preserve enzymatic activity

  • Inclusion of appropriate controls, including samples with known GSTA4 deficiency or overexpression

  • Normalization strategies accounting for total protein content or cell number

  • Validation using multiple methodological approaches

  • Consideration of potential inhibitors or activators present in biological samples

How can researchers effectively design experiments to investigate GSTA4's role in drug resistance?

Designing effective experiments to investigate GSTA4's role in drug resistance requires multifaceted approaches that address both mechanistic questions and clinical relevance. Based on current research, the following experimental design strategies are recommended:

  • Cell Line Models with Modulated GSTA4 Expression:

    • Generate isogenic cell lines with GSTA4 knockout, knockdown, and overexpression

    • Validate changes in GSTA4 expression at both mRNA and protein levels

    • Confirm functional changes using 4-HNE sensitivity assays

  • Dose-Response Studies:

    • Perform comprehensive dose-response experiments with clinically relevant chemotherapeutics (e.g., 5-FU, oxaliplatin)

    • Calculate IC50 values and compare between GSTA4-modulated and control cells

    • Assess time-dependent effects through exposure time course experiments

  • Mechanistic Investigations:

    • Measure drug-induced DNA damage using γH2AX quantification by flow cytometry or immunofluorescence

    • Assess ROS levels using multiple detection methods (e.g., DCFDA, MitoSOX)

    • Analyze activation of key signaling pathways (AKT, p38 MAPK) through phosphorylation status

    • Investigate apoptotic markers to determine cell death mechanisms

  • In Vivo Validation:

    • Establish xenograft models using GSTA4-modulated cancer cells

    • Administer chemotherapeutic agents at clinically relevant doses

    • Monitor tumor growth, drug response, and toxicity parameters

    • Perform immunohistochemical analysis of excised tumors for markers of proliferation, apoptosis, and DNA damage

  • Clinical Sample Analysis:

    • Correlate GSTA4 expression in patient samples with treatment outcomes

    • Stratify patients based on GSTA4 expression or polymorphisms

    • Analyze paired pre- and post-treatment samples for changes in GSTA4 expression

  • Combination Approaches:

    • Test GSTA4 inhibition or silencing in combination with standard chemotherapeutics

    • Investigate potential synergistic effects using Chou-Talalay method or similar approaches

    • Explore combinations with targeted therapies that affect pathways modulated by GSTA4

This comprehensive experimental approach allows researchers to establish both correlative and causative relationships between GSTA4 and drug resistance, providing insights that may ultimately inform clinical strategies for overcoming chemoresistance.

What emerging technologies could advance our understanding of GSTA4 biology?

Several emerging technologies hold significant promise for advancing our understanding of GSTA4 biology across multiple research dimensions:

  • CRISPR-Based Functional Genomics: Beyond simple gene knockout, CRISPR interference (CRISPRi) and CRISPR activation (CRISPRa) systems allow for fine-tuned modulation of GSTA4 expression . High-throughput CRISPR screens can identify synthetic lethal interactions with GSTA4, revealing new therapeutic vulnerabilities in GSTA4-overexpressing cancers.

  • Single-Cell Technologies: Single-cell RNA sequencing and proteomics can reveal cell-to-cell heterogeneity in GSTA4 expression within tumors, potentially identifying resistant subpopulations. These approaches may uncover previously unrecognized roles of GSTA4 in specific cellular subtypes.

  • Spatial Transcriptomics and Proteomics: These technologies can map GSTA4 expression patterns within the tumor microenvironment, providing insights into how GSTA4 expression varies spatially relative to other features such as hypoxia, immune infiltration, or stromal interactions.

  • Protein-Protein Interaction Mapping: Advanced techniques like BioID, APEX proximity labeling, or hydrogen-deuterium exchange mass spectrometry can identify direct interaction partners of GSTA4, potentially revealing new signaling functions beyond its enzymatic activity.

  • Organoid Models: Patient-derived organoids provide physiologically relevant 3D models for studying GSTA4 function in a context that better recapitulates tumor architecture and heterogeneity than traditional 2D cell culture.

  • In Situ GSTA4 Activity Probes: Development of fluorescent or bioluminescent probes that specifically detect GSTA4 activity in living cells could provide real-time visualization of how GSTA4 activity fluctuates in response to various stressors or treatments.

  • Systems Biology Approaches: Multi-omics integration combining transcriptomics, proteomics, metabolomics, and lipidomics can provide a comprehensive view of how GSTA4 influences cellular metabolism and signaling networks.

  • Structural Biology Advances: Cryo-electron microscopy and computational approaches like AlphaFold can provide detailed structural insights into GSTA4's interaction with substrates and potential binding partners, facilitating structure-based drug design.

Implementing these technologies will require interdisciplinary collaboration but promises to significantly expand our understanding of GSTA4's multifaceted roles in both normal physiology and disease states.

What are the current challenges and knowledge gaps in GSTA4 human research?

Despite significant advances in understanding GSTA4's role in cancer and other pathological conditions, several important challenges and knowledge gaps remain in GSTA4 human research:

Addressing these challenges will require multidisciplinary approaches combining biochemistry, molecular biology, structural biology, systems biology, and clinical research to develop a more comprehensive understanding of GSTA4 in human health and disease.

Product Science Overview

Introduction

Glutathione S-Transferase Alpha 4 (GSTA4) is an enzyme encoded by the GSTA4 gene in humans. It belongs to the family of glutathione S-transferases (GSTs), which are crucial for cellular detoxification processes. These enzymes catalyze the conjugation of reduced glutathione to a variety of hydrophobic and electrophilic compounds, aiding in their detoxification and excretion .

Gene and Protein Structure

The GSTA4 gene is located on chromosome 6 and is part of the alpha class of GSTs. This class of enzymes is characterized by their glutathione peroxidase activity, which is essential for detoxifying lipid peroxidation products . The GSTA4 protein is a homodimer, meaning it consists of two identical subunits, and it exhibits high catalytic efficiency with 4-hydroxyalkenals such as 4-hydroxynonenal (4-HNE) .

Function and Mechanism

GSTA4 plays a significant role in protecting cells from oxidative stress by detoxifying reactive electrophiles produced during oxidative metabolism. These reactive electrophiles have been linked to various degenerative diseases, including Parkinson’s disease, Alzheimer’s disease, cataract formation, and atherosclerosis . The enzyme’s ability to conjugate reduced glutathione to harmful compounds makes it a vital component of the cellular defense mechanism .

Clinical Significance

Mutations or alterations in the GSTA4 gene have been associated with several diseases. For instance, GSTA4 is linked to osteogenesis imperfecta type IV and cataracts . The enzyme’s role in detoxifying lipid peroxidation products also implicates it in conditions related to oxidative stress and inflammation .

Recombinant GSTA4

Recombinant human GSTA4 is produced using recombinant DNA technology, which involves inserting the human GSTA4 gene into a suitable expression system, such as bacteria or yeast, to produce the enzyme in large quantities. This recombinant enzyme is used in various research and clinical applications, including studies on detoxification mechanisms, drug metabolism, and the development of therapeutic interventions for diseases associated with oxidative stress .

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.