GPX7 Human

Glutathione Peroxidase 7 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of GPX7 Human

GPX7 (glutathione peroxidase 7), also known as non-selenocysteine glutathione peroxidase (NPGPx), is a cysteine-dependent peroxidase localized in the endoplasmic reticulum (ER) and secretory pathway. Unlike other GPX family members, it lacks selenocysteine and demonstrates minimal glutathione peroxidase activity . GPX7 functions as a redox sensor, transmitting oxidative stress signals to interacting proteins via disulfide bond shuttling .

PropertyDetail
Gene3 exons, 187 amino acids (22 kDa)
LocalizationER, Golgi apparatus (post-translational processing)
Catalytic SitesCys57 and Cys86 (disulfide bond formation)
Key InteractionsGRP78, PDI, TNFR1, TRAF2, Ero1α
Primary FunctionsOxidative stress sensing, protein folding, ER stress response, immune regulation

Functional Roles in Cellular Homeostasis

GPX7 regulates oxidative stress and ER protein folding through distinct mechanisms:

Oxidative Stress Sensing and Signal Transmission

  • GRP78 Activation: Oxidized GPX7 forms disulfide bonds with GRP78 (Cys41/Cys420), enhancing its chaperone activity and ER stress response .

  • PDI Oxidation: Promotes H₂O₂-dependent oxidation of protein disulfide isomerase (PDI), enabling disulfide bond formation during protein folding .

Table 2: GPX7’s Role in ER Stress and Protein Folding

ProcessMechanism
GRP78 ActivationDisulfide bond formation (Cys86 of GPX7 ↔ Cys41/Cys420 of GRP78)
PDI OxidationGPX7/Ero1α/PDI complex accelerates oxidative folding in ER
ROS RegulationModulates H₂O₂ levels to prevent excessive oxidative damage

Immune and Inflammatory Regulation

  • NF-κB Suppression: Loss of GPX7 enhances TNF-α-induced NF-κB activation by stabilizing TNFR1 and TRAF2 .

  • Immune Cell Infiltration: Overexpression in gliomas correlates with altered immune cell infiltration and type I interferon signaling .

Clinical and Prognostic Significance

GPX7’s dysregulation is implicated in multiple diseases, including cancers and metabolic disorders.

Oncological Roles

  • Gliomas: Elevated GPX7 expression predicts poor prognosis in glioblastoma (GBM) and low-grade glioma (LGG) .

  • Tumor Microenvironment: Modulates immune cell infiltration and ferroptosis resistance .

Table 3: GPX7’s Prognostic Value in Cancers

Cancer TypePrognostic ImpactSource
Glioblastoma (GBM)High expression → Poor OS (P < 0.001)
Low-Grade GliomaHigh expression → Poor DFS (P = 3.6e-05)
Breast CancerOverexpression → Resistance to oxidative stress

Epigenetic Regulation

  • DNA Methylation: Hypomethylation of the GPX7 promoter correlates with its overexpression in gliomas .

  • Histone Acetylation: Enrichment of H3K9ac/H3K27ac marks near the GPX7 promoter drives transcriptional activation .

Table 4: Epigenetic Mechanisms Regulating GPX7

MechanismEffect
HypomethylationUpregulates GPX7 expression in gliomas
H3K9ac/H3K27acMarks active chromatin regions, enhancing transcriptional activation

Ferroptosis Resistance

GPX7 knockdown increases lipid peroxidation and ferroptosis sensitivity in glioma cells, suggesting its role in tumor survival .

miRNA Regulation

  • miR-29b: Targets GPX7 mRNA, reducing its expression and enhancing ferroptosis .

Table 5: miRNA-GPX7 Interactions

miRNAEffect
miR-29bInhibits GPX7 translation, promoting ferroptosis in glioma

Future Directions and Research Gaps

  • Therapeutic Strategies: Targeting GPX7-PDI interactions or miR-29b-GPX7 axis may offer new approaches for glioma treatment .

  • Systemic Oxidative Stress: GPX7 knockout models show multi-organ damage, warranting further study in metabolic diseases .

Product Specs

Introduction
Glutathione peroxidase 7 (GPX7) is a member of the glutathione peroxidase family, a group of enzymes known for their peroxidase activity. These enzymes play a crucial role in protecting organisms from oxidative stress by reducing lipid hydroperoxides to their corresponding alcohols and converting free hydrogen peroxide to water.
Description
Recombinant human GPX7, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 193 amino acids (residues 20-187), resulting in a molecular weight of 21.8 kDa. The protein includes a 25 amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The GPX7 solution is provided at a concentration of 1 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 100 mM NaCl, 1 mM DTT, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), the solution can be stored at 4°C. For longer storage, freeze the solution at -20°C. Adding a carrier protein like 0.1% HSA or BSA is recommended for long-term storage. It's important to avoid repeated freezing and thawing of the solution.
Purity
The purity of the protein is confirmed to be greater than 95% using SDS-PAGE analysis.
Synonyms
Glutathione peroxidase 7, glutathione peroxidase 6, GPX6, NPGPx, CL683, GPx-7, GSHPx-7, non-selenocysteine containing phospholipid hydroperoxide glutathione peroxidase, FLJ14777, EC 1.11.1.9.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMQQEQD FYDFKAVNIR GKLVSLEKYR GSVSLVVNVA SECGFTDQHY RALQQLQRDL GPHHFNVLAF PCNQFGQQEP DSNKEIESFA RRTYSVSFPM FSKIAVTGTG AHPAFKYLAQ TSGKEPTWNF WKYLVAPDGK VVGAWDPTVS VEEVRPQITA LVRKLILLKR EDL

Q&A

What is GPX7 and what distinguishes it from other glutathione peroxidases?

GPX7 (Glutathione peroxidase 7) is a non-selenium containing enzyme that belongs to the glutathione peroxidase family. Unlike classic GPXs (GPX1-4) that contain selenocysteine, GPX7 utilizes a cysteine residue in its peroxidatic center . It is primarily localized to the endoplasmic reticulum (ER) where it plays crucial roles in oxidative protein folding and maintenance of ER redox control by eliminating H2O2 . GPX7 contains both a peroxidatic cysteine (CP) and a resolving cysteine (CR), with the latter regulating its protein disulfide isomerase (PDI) oxidation activity . A key structural feature that enhances GPX7's functionality is its catalytic tetrad at the redox-active site, which stabilizes the sulfenylated species generated upon reaction with H2O2 .

How does the reactivity of GPX7 compare to GPX8?

Experimental evidence demonstrates that GPX7 functions as a much more efficient PDI oxidase than GPX8 due to its higher susceptibility to H2O2 . This difference in reactivity is attributed to the presence of a glutamine residue (Gln92) in GPX7's catalytic tetrad, which is replaced by serine in GPX8 . When Gln92 in GPX7 is mutated to serine (GPX7 Q92S), its reactivity with H2O2 is greatly compromised, and its PDI oxidation activity is abolished . Interestingly, GPX7-mediated PDI oxidation is slower at higher H2O2 concentrations (200 μM) than at lower concentrations (50 μM), likely due to the formation of hyperoxidized GPX7 species during catalysis at higher H2O2 concentrations .

Which protein partners does GPX7 preferentially interact with in cells?

Research has revealed that GPX7 forms complexes preferentially with specific protein disulfide isomerase (PDI) family members, particularly PDI and P5, in H2O2-treated cells . This selective interaction suggests that GPX7 plays a targeted role in oxidizing specific PDI proteins involved in distinct protein folding pathways . The preferential binding to these partners appears to be functionally significant, as GPX7 efficiently catalyzes their oxidation, thereby facilitating proper disulfide bond formation in nascent proteins within the ER .

What techniques are most effective for measuring GPX7 enzymatic activity?

Several complementary approaches can be used to effectively measure GPX7 enzymatic activity:

  • PDI oxidation assay: Monitor the oxidation state of PDI in the presence of GPX7 and H2O2 using non-reducing SDS-PAGE to visualize migration patterns of reduced and oxidized PDI forms . This assay directly demonstrates GPX7's PDI oxidase activity.

  • H2O2 consumption assay: Measure the rate of H2O2 elimination in the presence of GPX7 using peroxide-sensitive probes or enzymatic coupled reactions.

  • Detection of oxidized GPX7 species: Analyze the formation of sulfenylated and hyperoxidized GPX7 species during catalysis using specific antibodies or mass spectrometry-based approaches .

When conducting these assays, it's crucial to carefully control H2O2 concentration, as research has shown that GPX7 activity varies significantly depending on H2O2 levels, with hyperoxidation occurring at higher concentrations (200 μM) .

How can researchers effectively study GPX7 in cancer models?

Effective approaches for studying GPX7 in cancer models include:

What methodological considerations are important when analyzing GPX7 regulation in different cellular contexts?

When analyzing GPX7 regulation across different cellular contexts, researchers should consider these methodological approaches:

  • Epigenetic regulation analysis:

    • DNA methylation profiling of the GPX7 promoter region

    • Histone modification analysis at the GPX7 locus

    • Treatment with epigenetic modifiers (HDAC inhibitors, DNA methyltransferase inhibitors) to assess impact on GPX7 expression

  • Cell cycle-dependent expression:

    • Synchronize cells at different cell cycle phases and analyze GPX7 expression

    • Use flow cytometry to correlate GPX7 expression with cell cycle markers

    • Assess impact of cell cycle inhibitors on GPX7 expression

  • Context-specific function assessment:

    • Compare GPX7 functions in different cell types (e.g., neuronal cells versus glioma cells)

    • Analyze pathway enrichment in different contexts (e.g., immune mechanisms in LGG versus metabolic regulation in GBM)

    • Consider microenvironmental factors that might influence GPX7 regulation (hypoxia, nutrient availability, oxidative stress)

  • Protein partner interactions:

    • Perform co-immunoprecipitation studies under different cellular conditions

    • Use proximity ligation assays to visualize context-dependent interactions

    • Apply FRET/BRET approaches to detect dynamic protein-protein interactions

  • Statistical validation:

    • Use multiple independent cohorts or datasets to validate findings

    • Apply appropriate multivariate analyses to account for confounding factors

    • Ensure sufficient statistical power for detecting context-specific differences

What is the prognostic significance of GPX7 in gliomas?

The prognostic significance of GPX7 in gliomas, particularly lower-grade gliomas (LGG), is substantial and well-documented:

These findings highlight GPX7's potential utility as both a prognostic biomarker and a predictor of treatment response in gliomas.

How does GPX7 dysregulation contribute to mechanisms of tumorigenesis?

GPX7 dysregulation contributes to tumorigenesis through multiple mechanisms:

What experimental evidence links GPX7 to metabolic diseases?

Several lines of experimental evidence link GPX7 to metabolic diseases:

  • Adipocyte differentiation: Studies in mice demonstrate that loss of GPX7 leads to increased differentiation of preadipocytes , suggesting a role in regulating adipose tissue development and potentially contributing to obesity.

  • Lipotoxicity prevention: GPX7 helps prevent palmitic acid-induced lipotoxicity . Palmitic acid is a saturated fatty acid that can induce cellular stress and inflammation, contributing to insulin resistance and other metabolic disorders.

  • Shortened lifespan: Loss of GPX7 in mice results in shortened lifespan , which may be partly due to accelerated development of age-related metabolic disorders.

  • Redox regulation: By maintaining redox homeostasis, GPX7 helps protect against oxidative stress-induced metabolic dysfunction. Oxidative stress is a key contributor to the development of metabolic diseases.

  • ER homeostasis: As an ER-resident enzyme, GPX7 contributes to ER homeostasis. ER stress is increasingly recognized as a central feature of metabolic diseases including obesity, diabetes, and non-alcoholic fatty liver disease.

These findings collectively suggest that GPX7 plays important roles in metabolic health, with dysregulation potentially contributing to the development of metabolic disorders.

How does the catalytic tetrad in GPX7 enhance its reactivity with H2O2?

The catalytic tetrad in GPX7 enhances its reactivity with H2O2 through several sophisticated mechanisms:

  • Structural stabilization: The catalytic tetrad, particularly the glutamine residue at position 92 (Gln92), stabilizes the sulfenylated species (CP-SOH) generated when the peroxidatic cysteine (CP) reacts with H2O2 . This stabilization prevents rapid, non-productive reactions and allows for controlled catalysis.

  • Experimental evidence: The critical role of Gln92 has been demonstrated through swap mutation experiments. When Gln92 in GPX7 is mutated to serine (GPX7 Q92S), the enzyme's reactivity with H2O2 is greatly compromised, and its PDI oxidation activity is abolished .

  • Microenvironment optimization: The structure of the catalytic tetrad creates an optimal microenvironment that increases the susceptibility of the peroxidatic cysteine to H2O2, making GPX7 more reactive than other similar enzymes like GPX8 .

  • Redox potential modulation: The catalytic tetrad likely influences the redox potential of the peroxidatic cysteine, lowering its pKa and making it more reactive at physiological pH.

  • Hyperoxidation protection: Despite enhancing reactivity with H2O2, the catalytic tetrad may also provide some protection against hyperoxidation at moderate H2O2 concentrations, although at higher H2O2 levels (200 μM), hyperoxidation still occurs .

This sophisticated catalytic mechanism makes GPX7 a highly efficient H2O2-dependent PDI oxidase with important implications for its biological functions.

What is the mechanism of GPX7-mediated protein disulfide isomerase (PDI) oxidation?

The mechanism of GPX7-mediated PDI oxidation involves a multi-step process:

  • Initial H2O2 reaction: The peroxidatic cysteine (CP) of GPX7 reacts with H2O2 to form a sulfenylated intermediate (CP-SOH) .

  • Resolving cysteine interaction: The resolving cysteine (CR) in GPX7 reacts with the sulfenylated CP to form an intramolecular disulfide bond, regulating GPX7's PDI oxidation activity .

  • PDI interaction: Oxidized GPX7 interacts with reduced PDI, transferring its disulfide bond to PDI's active site thiols.

  • Kinetic efficiency: GPX7 generates significant amounts of partially and fully oxidized PDI at early time points, whereas GPX8 hardly accelerates PDI oxidation compared with H2O2 alone .

  • H2O2 concentration dependence: GPX7-mediated PDI oxidation is slower at higher H2O2 concentrations (200 μM) than at lower concentrations (50 μM), likely due to the greater generation of hyperoxidized GPX7 species during catalysis .

  • Substrate specificity: GPX7 preferentially forms complexes with specific PDI family members, particularly PDI and P5, in H2O2-treated cells , suggesting targeted oxidation of specific PDI proteins involved in distinct protein folding pathways.

This mechanism allows GPX7 to function as an efficient H2O2-dependent PDI oxidase, facilitating oxidative protein folding in the endoplasmic reticulum.

How does GPX7 contribute to redox signaling networks in different cellular contexts?

GPX7 contributes to redox signaling networks in multiple ways across different cellular contexts:

  • ER redox environment regulation: By eliminating H2O2 in the endoplasmic reticulum, GPX7 helps maintain the optimal redox environment needed for proper protein folding . This regulation influences broader ER-dependent signaling pathways.

  • Context-specific pathway modulation:

    • In lower-grade gliomas (LGG), GPX7 is involved in immune mechanisms encompassing both innate and adaptive immunity, type I interferon production, and regulation of synaptic transmission .

    • In glioblastoma (GBM), GPX7 is primarily related to metabolic regulation of mitochondrial dynamics .

  • Immune signaling: GPX7 strongly correlates with immune cell infiltration and diverse immune cell markers in tumors , suggesting its involvement in redox-dependent immune signaling pathways.

  • Cell cycle regulation: GPX7 is expressed in a cell cycle-dependent manner , indicating potential involvement in redox-regulated cell cycle checkpoints.

  • Adaptive response to oxidative stress: By efficiently reacting with H2O2, GPX7 likely serves as a sensor of oxidative stress, translating changes in H2O2 levels into specific cellular responses.

  • Disulfide relay systems: Through its interactions with PDI family members, GPX7 participates in disulfide relay systems that can transmit redox signals throughout the cell.

This versatile involvement in different signaling networks highlights GPX7's role as a key node in cellular redox communication systems, contributing to context-specific cellular responses to changing redox conditions.

What are the most promising therapeutic opportunities targeting GPX7 in cancer?

Several promising therapeutic opportunities targeting GPX7 in cancer are emerging:

  • Biomarker-guided treatment selection: GPX7 expression shows significant negative correlation with temozolomide (TMZ) sensitivity in lower-grade gliomas (spearman= −0.59, P =1.3e-48) . This strong correlation suggests GPX7 could serve as a predictive biomarker for selecting patients most likely to benefit from TMZ therapy.

  • Combination therapy approaches:

    • For tumors with high GPX7 expression, combining standard therapies with agents that modulate redox balance may enhance efficacy

    • In TMZ-resistant cases with low GPX7 expression, alternative treatment strategies targeting other pathways might be preferred

  • Immunotherapy enhancement: Given GPX7's correlation with immune cell infiltration and immune markers , modulating GPX7 activity might enhance response to immunotherapies by altering the tumor immune microenvironment.

  • Epigenetic intervention: Since GPX7 upregulation in tumors is "tightly modulated by epigenetic processes" , epigenetic therapies that normalize GPX7 expression could offer therapeutic benefits.

  • Direct GPX7 inhibition: Developing small molecules that selectively inhibit GPX7's catalytic activity could be valuable for tumors where GPX7 overexpression drives progression.

  • Metabolic targeting: In glioblastoma, where GPX7 is mainly related to "metabolic regulation of mitochondrial dynamics" , combining GPX7 modulation with metabolic interventions might disrupt cancer cell energy production.

  • Prognostic stratification: Using GPX7 expression as part of a prognostic nomogram (C-index: 0.845; 95% CI, 0.825–0.865) could help identify high-risk patients who might benefit from more aggressive treatment approaches.

What methodological advances are needed to better understand GPX7's role in human disease?

Several methodological advances would significantly enhance our understanding of GPX7's role in human disease:

  • Improved protein activity assays:

    • Development of selective probes to monitor GPX7 activity in living cells

    • High-throughput screening methods to identify GPX7 modulators

    • Techniques to distinguish between GPX7 and GPX8 activities in complex biological samples

  • Advanced imaging approaches:

    • Super-resolution microscopy to visualize GPX7 localization and dynamics within the ER

    • FRET/BRET sensors to monitor GPX7-substrate interactions in real-time

    • In vivo imaging methods to track GPX7 activity in animal models

  • Systems biology integration:

    • Multi-omics approaches combining transcriptomics, proteomics, and metabolomics to understand GPX7's impact on cellular networks

    • Machine learning algorithms to identify GPX7-associated molecular signatures across diseases

    • Network analysis tools to map GPX7's position in redox signaling pathways

  • Translational research tools:

    • Biobanks with well-annotated clinical samples for GPX7 expression analysis

    • Patient-derived organoids to study GPX7 function in personalized disease models

    • Standardized immunohistochemistry protocols for consistent GPX7 detection in clinical samples

  • Genetic engineering advances:

    • Inducible, tissue-specific GPX7 knockout or overexpression systems

    • CRISPR-based approaches for precise modification of GPX7 catalytic residues

    • Humanized mouse models expressing human GPX7 variants

  • Computational methods:

    • Molecular dynamics simulations to understand GPX7 catalytic mechanisms

    • Structural biology approaches to design selective GPX7 modulators

    • Pharmacogenomic analyses to predict response to GPX7-targeting therapies

These methodological advances would collectively accelerate our understanding of GPX7's diverse roles in human disease and facilitate the development of GPX7-based diagnostic and therapeutic strategies.

How does current evidence suggest GPX7 involvement in neurological disorders?

Current evidence suggests several mechanisms of GPX7 involvement in neurological disorders:

  • Association with neurodegeneration: Research has established that "numerous pathological conditions, such as neurodegeneration, cancer and metabolic diseases have been linked with altered GPX7 and GPX8 expression" , indicating a recognized connection between GPX7 and neurodegenerative processes.

  • Synaptic transmission regulation: In lower-grade gliomas, GPX7 has been implicated in "regulation of synaptic transmission" , suggesting a potential role in neuronal function that could be relevant to various neurological disorders.

  • Protein folding and ER stress: GPX7's function in oxidative protein folding and ER homeostasis is particularly relevant to neurodegeneration, as protein misfolding and ER stress are hallmarks of conditions such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis.

  • Redox balance in neural cells: The brain is especially vulnerable to oxidative stress due to its high oxygen consumption and relatively limited antioxidant capacity. GPX7's role in maintaining redox homeostasis could be critical for neuronal survival and function.

  • Immune modulation: GPX7's involvement in immune mechanisms may be relevant to neuroinflammatory aspects of neurological disorders, as neuroinflammation is increasingly recognized as a contributor to neurodegenerative and neuroimmune diseases.

  • Mitochondrial regulation: The finding that GPX7 is related to "metabolic regulation of mitochondrial dynamics" in glioblastoma suggests potential involvement in mitochondrial function, which is frequently compromised in neurological disorders.

While direct studies of GPX7 in specific neurological disorders are still emerging, these mechanistic connections provide compelling evidence for its potential involvement and highlight the need for further investigation in this area.

Product Science Overview

Gene and Protein Structure

The GPX7 gene is located on chromosome 1 at the 1p32.3 region . The gene contains three distinct introns and produces four different mRNA transcripts through alternative splicing . The protein itself is a non-selenocysteine-containing enzyme, which differentiates it from other members of the glutathione peroxidase family .

The recombinant human GPX7 protein is typically produced in E. coli or HEK293 cells. It is a single, non-glycosylated polypeptide chain containing 193 amino acids, with a molecular mass of approximately 21.8 kDa . The recombinant protein is often fused with tags such as His-tag or Fc region of human IgG1 to facilitate purification and detection .

Function and Mechanism

GPX7 exhibits glutathione peroxidase activity, which involves the reduction of hydrogen peroxide and organic hydroperoxides using glutathione as a substrate . This activity is crucial for cellular defense against oxidative stress, which can cause damage to proteins, lipids, and DNA. GPX7 is also involved in the metabolism of arachidonic acid and glutathione .

Biological Significance

GPX7 is expressed in various tissues, including the extracellular space and endoplasmic reticulum . It plays a significant role in cellular responses to oxidative stress and detoxification of cellular oxidants . The enzyme has been implicated in several physiological and pathological processes, including bone turnover and response to oxidative stress .

Clinical Relevance

Mutations or dysregulation of the GPX7 gene have been associated with certain diseases, such as amnestic disorder and Keshan disease . Additionally, GPX7 has been studied for its potential role in cancer, particularly in alleviating oxidative stress in breast cancer cells .

Applications

Recombinant human GPX7 is widely used in research to study its function and role in various biological processes. It is also utilized in drug development and therapeutic applications aimed at mitigating oxidative stress-related damage .

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.