GPX1 Human

Glutathione Peroxidase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

GPX1 Human Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 223 amino acids (1-203) and having a molecular mass of 24.2kDa.
GPX1 is fused to a 20 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Glutathione peroxidase 1 (GPX1) is a crucial antioxidant enzyme in humans, belonging to a family of eight glutathione peroxidases (Gpx1-8). It plays a vital role in detoxifying hydrogen peroxide, protecting DNA, proteins, and lipids from oxidative damage. This protection is significant as hydrogen and lipid peroxides can contribute to prostate cancer development. GPX1 is unique as it's one of the few proteins in higher vertebrates containing selenocysteine at its active site, encoded by the stop codon TGA. Additionally, GPX1 exhibits polymorphism in a polyalanine sequence within its N-terminal region. This polymorphism involves alleles with 5, 6, or 7 alanine repeats, with the 5 ALA repeat allele linked to breast cancer risk.
Description
Recombinant human GPX1, produced in E. coli, is a non-glycosylated polypeptide chain consisting of 223 amino acids (1-203). It has a molecular weight of 24.2kDa, achieved by fusing a 20 amino acid His-tag at the N-terminus and purifying it using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The GPX1 solution is prepared at a concentration of 0.5mg/ml in a buffer containing 20mM Tris-HCl (pH 8.0), 2mM DTT, 30% glycerol, and 100mM NaCl.
Stability
For short-term storage (up to 4 weeks), the GPX1 solution can be stored at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. Adding a carrier protein like 0.1% HSA or BSA is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of GPX1 is determined to be greater than 90% using SDS-PAGE analysis.
Synonyms
Glutathione peroxidase 1, GPx-1, GSHPx-1, Cellular glutathione peroxidase, GPX1, GPXD, GSHPX1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MCAARLAAAA AAAQSVYAFS ARPLAGGEPV SLGSLRGKVL LIENVASLCG TTVRDYTQMN ELQRRLGPRG LVVLGFPCNQ FGHQENAKNE EILNSLKYVR PGGGFEPNFM LFEKCEVNGA GAHPLFAFLR EALPAPSDDA TALMTDPKLI TWSPVCRNDV AWNFEKFLVG PDGVPLRRYS RRFQTIDIEP DIEALLSQGP SCA.

Q&A

What is the molecular structure of GPX1 and how does it function in human cells?

GPX1 is encoded by the GPX1 gene located on chromosome 3 and forms a homotetramer structure with four homogenous subunits, each containing one selenocysteine residue . This enzyme functions as part of the glutathione peroxidase family, consisting of eight known glutathione peroxidases (GPx1-8) in humans .

GPX1's catalytic mechanism involves the reduction of H₂O₂ and soluble low-molecular hydroperoxides. During this reaction, the selenol (SE-H) active site is oxidized to selenic acid (SE-OH), which is subsequently reduced by glutathione (GSH) to form a glutathioneated selenol (SE-SG) intermediate . A second GSH molecule further reduces this intermediate, forming oxidized glutathione (GSSG) and restoring the active site, with NADPH-dependent glutathione reductase completing the redox cycle .

How is GPX1 distributed within human tissues and cells?

GPX1 is ubiquitously expressed in many tissues throughout the human body, where it protects cells from oxidative stress . Within cells, GPX1 primarily localizes to the cytoplasm and mitochondria . Research has shown that high levels of GPX1 are present in tissues with high oxidative metabolism, such as red blood cells, placenta, lung, liver, and kidney .

What are the standard techniques for studying GPX1 polymorphisms?

Researchers typically employ polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis to investigate GPX1 polymorphisms . This method is particularly useful for large-scale studies with limited DNA availability .

For the common polymorphisms, such as rs1800668 C/T and rs1050450 C/T, researchers design specific primers to amplify the target regions containing these variants, followed by restriction enzyme digestion that differentially cuts the DNA based on the presence of specific nucleotides . The resulting fragments are then analyzed by gel electrophoresis to determine the genotype.

The methodology typically involves:

  • DNA extraction from patient samples

  • PCR amplification of the target region

  • Restriction enzyme digestion

  • Fragment analysis by gel electrophoresis

  • Statistical analysis to determine associations with disease or phenotype

How can GPX1 expression be effectively measured in experimental and clinical samples?

Multiple complementary approaches are employed to comprehensively assess GPX1 expression:

  • mRNA Expression Analysis:

    • Quantitative real-time PCR for relative quantification

    • RNA sequencing for genome-wide expression profiling

  • Protein Expression Analysis:

    • Western blotting for semi-quantitative protein detection

    • Immunohistochemistry for tissue localization and expression patterns

    • ELISA for quantitative protein measurement

  • Database Analysis:

    • Platforms such as Oncomine and GEPIA provide tools to analyze GPX1 expression across thousands of samples from TCGA and GTEx projects

    • These databases allow comparison between tumor and normal tissues across multiple cancer types

For example, studies have employed GEPIA to demonstrate that GPX1 expression is significantly higher in glioblastoma multiforme, kidney renal papillary cell carcinoma, acute myeloid leukemia, and several other cancer types compared to adjacent normal tissues .

Which GPX1 polymorphisms have been linked to human diseases and what are their functional implications?

Several key polymorphisms in the GPX1 gene have been associated with various human diseases:

  • Pro198Leu Polymorphism (rs1050450 C/T):

    • Significantly associated with schizophrenia in Chinese Han populations

    • Studies show individuals with the CT genotype have a higher risk (OR=1.453, 95% CI: 1.055-2.002, p=0.021) compared to those with the CC genotype

    • Also linked to increased risk of lung, breast, head and neck, and bladder cancers

  • Promoter Region Polymorphism (rs1800668 C/T):

    • Often studied in conjunction with rs1050450

    • Forms significant haplotypes that show strong disease associations

  • Polyalanine Sequence Polymorphism:

    • Characterized by three alleles with five, six, or seven alanine repeats in the N-terminal region

    • The allele with five alanine repeats is significantly associated with increased breast cancer risk

HaplotypeTotal FrequencyCase Group RatioControl Group RatioOR95% CI
CC0.88183.6%91.9%0.4500.328-0.167
CT0.0346.4%0.9%7.1673.362-15.279
TC0.0326.7%0.2%30.0127.313-123.175
TT0.0533.3%6.9%0.4660.287-0.765

Table: Haplotype analysis of rs1800668 and rs1050450 polymorphisms in schizophrenia case-control study

How should researchers design case-control studies to investigate GPX1 polymorphisms in disease?

When designing case-control studies for GPX1 polymorphisms, researchers should consider:

  • Sample Size Determination:

    • Power calculations based on expected effect sizes and polymorphism frequencies

    • Adequate representation of diverse populations to account for ethnic variations

  • Patient Selection and Matching:

    • Clear diagnostic criteria (e.g., DSM-IV for psychiatric disorders)

    • Matching controls for age, sex, ethnicity, and relevant environmental exposures

    • Detailed exclusion criteria for both cases and controls

  • Genotyping Approach:

    • PCR-RFLP remains a reliable and cost-effective method

    • Consider multiple polymorphisms to enable haplotype analysis

    • Include quality control measures such as duplicate testing

  • Statistical Analysis:

    • Test multiple genetic models (dominant, recessive, additive)

    • Perform haplotype analysis to identify combined effects of multiple polymorphisms

    • Adjust for multiple testing and potential confounders

  • Functional Validation:

    • Correlate genotypes with enzyme activity or expression levels

    • Consider in vitro functional studies to elucidate mechanisms

How does GPX1 deficiency impact cellular response to oxidative stress?

GPX1 deficiency has profound effects on cellular responses to oxidative stress, with significant implications for tissue damage and disease progression:

What are the paradoxical effects of GPX1 in inflammatory responses and how should researchers interpret these findings?

Interestingly, GPX1 demonstrates seemingly contradictory effects in certain inflammatory contexts:

  • Pro-inflammatory Effects:

    • In acute lung injury models, GPX1 knockout mice surprisingly showed decreased LPS-mediated NFκB activation and reduced pro-inflammatory cytokine production in neutrophils

    • GPX1 knockout bronchoalveolar lavage fluid contained lower levels of LPS-mediators and fewer macrophages

    • These findings suggest that GPX1 may enhance initial inflammatory responses by promoting pro-inflammatory cytokine production in some contexts

  • Opposing Effects in Different Cell Types:

    • GPX1 knockout endothelial cells show higher adhesion molecule expression in response to LPS

    • Conversely, GPX1 overexpression in endothelial cells decreases adhesion molecule expression

  • Interpretation Challenges:

    • These contradictory findings may relate to the dual role of H₂O₂ as both a damaging molecule and an important signaling mediator

    • H₂O₂ can inhibit acute excessive inflammatory responses in some contexts

    • Different experimental conditions, including varying doses of inflammatory stimuli and observation timepoints, may contribute to these disparate findings

Researchers should address these paradoxes by:

  • Carefully specifying experimental conditions

  • Examining multiple timepoints to distinguish between acute and chronic effects

  • Investigating cell-type specific responses

  • Measuring both inflammatory markers and oxidative stress parameters

  • Considering the complex interplay between redox signaling and inflammatory pathways

How does GPX1 expression vary across different cancer types and what are the implications for cancer biology?

GPX1 expression shows remarkable variability across cancer types, with important implications for tumorigenesis and progression:

Cancer TypeGPX1 Expression vs. Normal TissuePotential Implications
Cervical cancerIncreasedPotential role in tumor progression
Gastric cancerIncreasedMay contribute to treatment resistance
Kidney cancerIncreasedAltered redox homeostasis
LymphomaVariable (increased in some types)Context-dependent effects
LeukemiaIncreasedMay promote survival of cancer cells
Lung cancerDecreasedPossible tumor suppressor role
SarcomaDecreasedAltered oxidative stress response
GlioblastomaIncreasedPotential therapeutic target
Ovarian cancerIncreasedRole in tumor microenvironment
Pancreatic cancerIncreasedMay support tumor metabolism

Analysis of RNA sequencing data from thousands of samples in the TCGA and GTEx projects confirms that GPX1 expression is significantly higher in glioblastoma multiforme, kidney renal papillary cell carcinoma, acute myeloid leukemia, brain lower grade glioma, ovarian serous cystadenocarcinoma, pancreatic adenocarcinoma, skin cutaneous melanoma, testicular germ cell tumors, thyroid carcinoma, and uterine corpus endometrial carcinoma compared to normal tissues .

These expression patterns suggest that GPX1 may play diverse roles in different cancer contexts:

  • In some cancers, increased GPX1 may help cancer cells cope with elevated oxidative stress

  • In others, decreased GPX1 may contribute to genomic instability through increased ROS damage

  • The specific impact likely depends on the tumor microenvironment, metabolic state, and genetic background

What experimental approaches can researchers use to evaluate GPX1 as a potential biomarker or therapeutic target in cancer?

To evaluate GPX1's potential as a cancer biomarker or therapeutic target, researchers should implement a multi-faceted experimental approach:

  • Expression Profiling:

    • Immunohistochemistry on tissue microarrays to assess protein expression across tumor stages

    • qRT-PCR and Western blot analysis for quantitative assessment

    • Utilization of public databases like Oncomine and GEPIA for large-scale expression analysis

    • Single-cell RNA sequencing to understand cellular heterogeneity within tumors

  • Survival Analysis:

    • Kaplan-Meier analysis to correlate GPX1 expression with patient outcomes

    • Multivariate Cox regression to determine independent prognostic value

    • Stratification by tumor molecular subtypes and treatment regimens

  • Functional Studies:

    • CRISPR/Cas9-mediated knockout or siRNA knockdown in cancer cell lines

    • Overexpression systems to assess oncogenic potential

    • Assessment of proliferation, migration, invasion, and apoptosis

    • Xenograft models to evaluate in vivo effects of GPX1 modulation

  • Therapeutic Targeting:

    • Small molecule screening for GPX1 inhibitors

    • Evaluation of synthetic lethality with existing cancer therapies

    • Assessment of combination approaches with other redox-modulating agents

    • Development of nanoparticle-based delivery systems for tissue-specific targeting

  • Biomarker Validation:

    • Prospective clinical studies with standardized measurement protocols

    • Integration with other established biomarkers

    • Analysis in liquid biopsies (circulating tumor cells, cell-free DNA)

    • Development of clinically applicable assays with robust reproducibility

How does selenium availability affect GPX1 expression and activity in experimental systems?

Selenium availability critically impacts GPX1 expression and activity through several mechanisms:

What methodological approaches should researchers use to study selenium-GPX1 interactions in human populations?

To effectively study selenium-GPX1 interactions in human populations, researchers should employ the following methodological approaches:

  • Selenium Status Assessment:

    • Measure plasma/serum selenium concentration as a biomarker of status

    • Consider selenoprotein P levels as a functional marker of selenium status

    • Analyze toenail or hair selenium for long-term exposure assessment

    • Account for geographical variations in selenium intake

  • GPX1 Functional Analysis:

    • Measure GPX1 activity in erythrocytes or platelets using standardized assays

    • Assess GPX1 protein levels through immunological methods

    • Quantify GPX1 mRNA expression in accessible tissues

  • Genotyping:

    • Screen for functional GPX1 polymorphisms, particularly Pro198Leu (rs1050450)

    • Consider polymorphisms in other genes involved in selenium metabolism

    • Perform haplotype analysis to identify combined genetic effects

  • Study Design Considerations:

    • Conduct case-control studies stratified by selenium status

    • Implement prospective cohort studies with baseline selenium measurement

    • Design randomized controlled trials of selenium supplementation with GPX1 activity as an outcome

    • Account for confounding factors like smoking, alcohol consumption, and dietary antioxidant intake

  • Advanced Biomarker Integration:

    • Measure multiple oxidative stress biomarkers (8-OHdG, MDA, isoprostanes)

    • Assess inflammatory markers alongside GPX1 and selenium status

    • Consider metabolomic profiling to identify selenium-responsive metabolic pathways

What strategies should researchers employ when designing GPX1 knockout or overexpression models?

When designing GPX1 genetic modification models, researchers should consider:

  • Model Selection:

    • Cell line models for mechanistic studies (HEK293, HepG2, disease-specific cell lines)

    • Primary cell cultures for physiological relevance

    • Transgenic mouse models for systemic effects

    • Conditional knockout systems for tissue-specific studies

  • Technology Considerations:

    • CRISPR/Cas9 for precise gene editing and complete knockout

    • siRNA/shRNA for transient or stable knockdown

    • Lentiviral/retroviral vectors for stable overexpression

    • Inducible expression systems to control timing and level of expression

  • Control Mechanisms:

    • Include wild-type controls and empty vector controls

    • For selenium studies, pair with selenium-supplemented and selenium-deficient conditions

    • Consider rescue experiments to confirm specificity

  • Validation Strategies:

    • Confirm genetic modification at DNA level (sequencing)

    • Verify changes in mRNA expression (qRT-PCR)

    • Assess protein levels (Western blot, immunocytochemistry)

    • Measure enzymatic activity to confirm functional impact

  • Experimental Design:

    • Include oxidative stress challenges (H₂O₂, paraquat, etc.)

    • Examine both basal conditions and stress responses

    • Investigate both acute and chronic effects

    • Consider compensatory mechanisms by assessing other antioxidant enzymes

  • Physiological Relevance:

    • For mouse models, recognize that GPX1 knockout mice develop normally under standard conditions

    • Challenge models with oxidative stressors to reveal phenotypes

    • Consider tissue-specific effects, particularly in tissues with high oxidative metabolism

How can researchers effectively integrate GPX1 research with broader redox biology and systems biology approaches?

Integrating GPX1 research within broader biological contexts requires sophisticated approaches:

  • Multi-omics Integration:

    • Combine transcriptomics, proteomics, and metabolomics data

    • Analyze GPX1-dependent changes in redox-sensitive proteins via redox proteomics

    • Use phosphoproteomics to identify redox-regulated signaling pathways

    • Apply metabolomics to identify alterations in redox-sensitive metabolic pathways

  • Network Analysis:

    • Construct protein-protein interaction networks centered on GPX1

    • Identify transcription factor networks regulated by GPX1-dependent redox changes

    • Apply pathway enrichment analysis to GPX1-dependent gene expression changes

    • Develop computational models of glutathione/GSH biochemistry

  • Advanced Imaging Techniques:

    • Use redox-sensitive fluorescent proteins to visualize real-time redox changes

    • Apply super-resolution microscopy to determine subcellular localization

    • Implement FRET-based sensors to monitor protein-protein interactions

    • Employ in vivo imaging in animal models to track tissue-specific responses

  • Functional Genomics:

    • Conduct genome-wide CRISPR screens to identify synthetic lethal interactions with GPX1

    • Perform ChIP-seq to identify redox-sensitive transcription factor binding sites

    • Use ATAC-seq to assess chromatin accessibility changes in response to altered GPX1 activity

    • Apply RNA-seq to characterize global transcriptional responses

  • Clinical Translation:

    • Correlate GPX1 genetic variants with clinical phenotypes across diseases

    • Develop predictive models incorporating GPX1 status and oxidative stress markers

    • Design personalized intervention strategies based on GPX1 genotype

    • Investigate GPX1 biomarkers in accessible patient samples (blood, urine, etc.)

Product Science Overview

Structure and Function

GPx1 is the most abundant version of glutathione peroxidase, found in the cytoplasm of nearly all mammalian tissues . The enzyme’s primary function is to detoxify hydrogen peroxide (H₂O₂) by reducing it to water (H₂O), using glutathione (GSH) as a substrate. This reaction is crucial for maintaining cellular redox balance and protecting cells from oxidative stress .

Recombinant Human GPx1

Recombinant Human Glutathione Peroxidase 1 is a human full-length protein, typically expressed in Escherichia coli (E. coli) for research purposes . The recombinant form is often tagged with a His-tag at the N-terminus to facilitate purification and detection . It is used in various applications, including SDS-PAGE and mass spectrometry (MS), and is typically purified to over 90% purity .

Biological Importance

GPx1 plays a vital role in protecting hemoglobin in erythrocytes (red blood cells) from oxidative breakdown . In platelets, it is involved in the metabolism of arachidonic acid, which is essential for the production of signaling molecules called eicosanoids . These functions highlight the enzyme’s importance in maintaining cellular integrity and function under oxidative stress conditions.

Clinical Relevance

The enzyme’s ability to detoxify hydrogen peroxide and lipid peroxides makes it a critical component of the body’s antioxidant defense system. It helps prevent oxidative damage to DNA, proteins, and lipids, which can contribute to various diseases, including cancer . GPx1 is one of the few proteins in higher vertebrates that contain selenocysteine at its active site, which is encoded by the unusual stop codon TGA .

Research Applications

Recombinant Human GPx1 is widely used in research to study its structure, function, and role in oxidative stress. It is also used in high-throughput screening assays to identify potential therapeutic compounds that can modulate its activity .

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.