GLRX Mouse

Glutaredoxin Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Characterization of Mouse GLRX Protein

Mouse GLRX is a 12–14.3 kDa cytoplasmic enzyme encoded by the Glrx gene (UniProt: Q9QUH0). Key features include:

PropertyDetails
Amino Acid Sequence107 residues (1-107aa) with N-terminal His-tag in recombinant forms
Active SiteConserved Cys-Pro-Tyr-Cys motif enabling thiol-disulfide exchange
Post-Translational ModificationsFive redox-sensitive cysteines (C23, C26, C79, C82, C208)
Recombinant ExpressionProduced in E. coli with >95% purity; stored in MES buffer (pH 5.0)

Metabolic Dysregulation

  • Glrx⁻/⁻ mice develop spontaneous fatty liver, hyperlipidemia, and insulin resistance by 8 months of age, even on a normal diet .

  • Increased S-glutathionylation of Sirtuin 1 (SIRT1) in these mice disrupts lipid metabolism, elevating LDL/VLDL cholesterol .

Immune and Inflammatory Responses

  • Glrx deletion in macrophages reduces LPS-induced IL-33 production by 50%, impairing NF-κB signaling .

  • Overexpression of GLRX in endothelial cells inhibits tumor angiogenesis but paradoxically accelerates tumor growth by altering redox signaling .

Liver Fibrosis

  • Aged Glrx⁻/⁻ mice exhibit elevated αSMA and cleaved caspase-3, indicating hepatic apoptosis and fibrosis .

  • Adeno-associated virus (AAV)-mediated Glrx delivery reduces collagen deposition by 40% in diet-induced NASH models .

Lung Cancer

  • Glrx deficiency increases S-glutathionylation of OTUB1, stabilizing the cystine transporter SLC7A11 and elevating glutathione (GSH) levels .

  • TCGA data show reduced GLRX expression in human lung adenocarcinoma (LUAD), correlating with poor prognosis .

Antioxidant Therapy

  • Intraperitoneal administration of recombinant GLRX attenuates age-related liver fibrosis by reducing TNF-α and TGF-β1 expression .

  • In allergen-challenged mice, GLRX overexpression diminishes airway inflammation via IL-33/ST2 pathway regulation .

Cancer

  • Pharmacological inhibition of GLRX in Krasᴳ¹²ᴰ-driven LUAD models reduces tumor burden by 30% through SLC7A11 destabilization .

Key Experimental Models and Reagents

Reagent/ModelApplicationSource
Recombinant Mouse GLRXSDS-PAGE, enzyme activity assaysBoster Bio
Anti-GLRX Antibody (AF3399)Western blot, immunohistochemistryR&D Systems
Glrx siRNAMacrophage redox signaling studiesPLOS ONE
AAV-Hep-GlrxLiver-specific gene therapyMDPI

Current Research Gaps

  • The role of GLRX in non-canonical redox pathways (e.g., Ubiquitin-OTUB1 interactions) remains poorly understood .

  • Long-term safety profiles of GLRX-based therapies require further validation in primate models .

Product Specs

Introduction
GLRX1, also known as Glutaredoxin 1, is an enzyme with multifaceted roles in cellular redox regulation. It exhibits glutathione-disulfide oxidoreductase activity, utilizing NADPH and glutathione reductase to reduce disulfide bonds in various molecules, including low molecular weight disulfides and proteins. This activity is crucial for maintaining cellular redox balance and protecting against oxidative stress. As a glutathione-dependent hydrogen donor for ribonucleotide reductase, GLRX1 participates in DNA synthesis by supporting the reduction of ribonucleotides to deoxyribonucleotides. Moreover, it contributes to the detoxification of reactive oxygen species (ROS) by directly reducing hydroperoxides and repairing ROS-mediated damage, highlighting its significance in cellular defense mechanisms against oxidative stress.
Description
This product consists of recombinant GLRX protein derived from mouse and produced in E. coli. It is engineered as a single, non-glycosylated polypeptide chain encompassing 130 amino acids, including the 107 amino acids of the GLRX protein (1-107 a.a) and a 23 amino acid His-tag fused at the N-terminus. The molecular weight of the recombinant protein is 14.3 kDa. Purification is achieved through proprietary chromatographic techniques to ensure high purity.
Physical Appearance
The product appears as a clear solution that has been sterilized by filtration.
Formulation
The GLRX protein is supplied in a solution with a concentration of 0.5 mg/ml. The solution is buffered with 20mM MES at pH 5.0 and contains 20% glycerol and 0.1M NaCl for stability.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. To further enhance stability during long-term storage, the addition of a carrier protein such as HSA or BSA (0.1%) is advisable. It is crucial to avoid repeated freeze-thaw cycles to preserve protein integrity.
Purity
The purity of the GLRX protein is greater than 95.0%, as determined by SDS-PAGE analysis.
Synonyms

Glutaredoxin-1, Thioltransferase-1, TTase-1, Glrx, Glrx1, Grx, Grx1.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSMAQEFVN CKIQSGKVVV FIKPTCPYCR KTQEILSQLP FKQGLLEFVD ITATNNTSAI QDYLQQLTGA RTVPRVFIGK DCIGGCSDLI SMQQTGELMT RLKQIGALQL

Q&A

What is GLRX in mice and what are its key functions?

GLRX (also known as GRX, GRX1, or Thioltransferase) is a small thiol-disulfide oxidoreductase enzyme that plays a crucial role in cellular redox homeostasis in mice. Mouse GLRX (Uniprot ID: Q9QUH0) primarily catalyzes the reduction of protein-glutathione mixed disulfides (protein deglutathionylation), using glutathione (GSH) as a cofactor. This approximately 12 kDa protein functions within a network of antioxidant enzymes to protect cells against oxidative stress .

Functionally, GLRX participates in diverse cellular processes including:

  • Protein deglutathionylation during oxidative stress responses

  • Regulation of redox-sensitive transcription factors

  • Modulation of inflammatory signaling pathways

  • Protection against oxidative damage in various tissues

The GLRX system is particularly important during infection and inflammation, where it helps regulate the balance between protective immune responses and excessive inflammatory damage.

How does GLRX expression vary across mouse tissues?

GLRX exhibits tissue-specific expression patterns in mice, with highest expression typically observed in metabolically active organs. Expression levels vary significantly by tissue type:

  • High expression: Liver, kidney, heart, and lung

  • Moderate expression: Spleen, skeletal muscle

  • Lower expression: Brain and nervous tissue

This differential expression must be considered when designing experimental protocols, as baseline levels will affect detection sensitivity requirements. Furthermore, within individual tissues, expression can vary by cell type, making single-cell analysis approaches particularly valuable for heterogeneous tissues.

When analyzing GLRX across different mouse tissues, researchers should standardize protein loading based on tissue-specific expression levels, often requiring different dilution factors for accurate quantification .

What are the most reliable methods for detecting GLRX in mouse samples?

Several complementary techniques provide robust detection of mouse GLRX:

Western Blot Analysis: Using specific antibodies such as the Human/Mouse Glutaredoxin 1/GLRX1 Antibody, GLRX can be detected at approximately 12 kDa under reducing conditions. This approach is suitable for relative quantification and has been validated across multiple mouse cell lines including NIH-3T3 and C2C12 .

ELISA: Sandwich enzyme immunoassays provide more precise quantification of GLRX protein levels. Commercial mouse GLRX ELISA kits typically offer:

  • Detection range: 78.13-5000 pg/mL

  • Sensitivity: ~35 pg/mL

  • Sample compatibility: Serum, plasma, tissue homogenates, and other biological fluids

Enzymatic Activity Assays: These measure functional GLRX activity rather than protein quantity, providing complementary information about catalytic capacity. Standard substrates include hydroxyethyl disulfide (HEDS) or protein-SSG substrates.

For comprehensive analysis, combining protein quantification with activity measurements provides the most complete understanding of GLRX status in experimental models.

What sample preparation considerations are crucial for GLRX analysis in mouse tissues?

Reliable GLRX analysis depends on careful sample preparation:

Preservation of Redox State: GLRX is highly sensitive to oxidation during sample processing. Include reducing agents (DTT or β-mercaptoethanol) in lysis buffers to maintain native redox state. For studies of glutathionylated proteins, include alkylating agents (N-ethylmaleimide) to prevent artificial oxidation during preparation.

Tissue-Specific Extraction: Different mouse tissues require adapted protocols:

  • For soft tissues (liver, kidney): Standard RIPA buffer with protease inhibitors is typically sufficient

  • For fibrous tissues (heart, skeletal muscle): More rigorous homogenization and possibly specialized extraction buffers may be needed

Sample Dilution Strategy: As documented in validation studies, different sample types require different dilution approaches:

  • Serum samples: 86% average recovery with dilutions between 1:2 and 1:16

  • EDTA plasma: 97% average recovery

  • Heparin plasma: 92% average recovery

Storage Conditions: Store processed samples at -80°C, and avoid repeated freeze-thaw cycles which dramatically reduce GLRX detection. For antibodies, store reconstituted reagents according to manufacturer specifications (typically 1 month at 2-8°C or 6 months at -20 to -70°C) .

How do I determine the optimal dilution factor for mouse GLRX ELISA?

Determining the optimal dilution factor for mouse GLRX ELISA requires systematic evaluation:

  • Preliminary Range Testing: Test representative samples at multiple dilutions (1:2, 1:4, 1:8, 1:16) to identify the appropriate range for your sample type.

  • Linearity Assessment: Plot the calculated concentration versus dilution factor for each sample. Valid dilutions will produce a linear relationship when adjusted for dilution factor. Non-linearity suggests matrix interference effects.

  • Standard Curve Positioning: Select dilutions that produce readings in the mid-portion of the standard curve (approximately 625-1250 pg/mL based on typical standard curves), where precision is highest. As shown in validation data, the optical density readings for standard concentrations typically follow a consistent pattern:

Concentration (pg/mL)ODCorrected OD
5000.001.9941.895
2500.001.5591.460
1250.001.2171.118
625.001.0370.938
312.500.6660.567
156.250.4150.316
78.130.2350.136
0.000.0990.000
  • Sample-Specific Optimization: Different sample types typically require different dilution strategies based on GLRX concentration and matrix effects.

  • Validation Through Recovery: Confirm accuracy at selected dilutions through spike-recovery experiments. Acceptable recovery rates should fall between 80-120% of expected values.

What controls are essential when studying GLRX in inflammatory response models?

When investigating GLRX in mouse inflammatory response models, several controls are essential:

Vehicle Controls: For studies using inflammatory stimuli like LPS, vehicle-treated controls must undergo identical handling and injection procedures. This is particularly important as LPS dose responses vary between species - for example, in some studies, doses that were twice or half those given to deermice produced indistinguishable responses in rats .

Time-Matched Controls: GLRX expression fluctuates with circadian rhythms and stress responses. Include time-matched controls for each experimental time point rather than a single control group.

Parallel Inflammatory Markers: Include well-characterized inflammatory markers (IL-6, TNF-α) to confirm successful inflammatory induction and contextualize GLRX changes within the broader inflammatory response.

Pathway Inhibitor Controls: Include specific inhibitors of inflammatory signaling pathways (NF-κB inhibitors) to delineate mechanisms regulating GLRX during inflammation.

Species Comparison Controls: Consider species differences in inflammatory responses. As demonstrated in comparative studies, white-footed deermice (Peromyscus leucopus) respond differently to inflammatory stimuli compared to laboratory mice and rats, particularly in endogenous retrovirus activation patterns .

Tissue-Specific Controls: Compare GLRX responses across different tissues, as the regulation of redox systems during inflammation exhibits tissue specificity.

How should I design experiments to study GLRX regulation in mouse models?

Robust experimental design for GLRX studies should address several key considerations:

Mouse Strain Selection: Different strains have distinct baseline GLRX expression and redox regulation. C57BL/6 and BALB/c mice have well-characterized redox profiles but may respond differently to the same stimulus.

Age and Sex Stratification: GLRX expression changes with age and differs between males and females. Design experiments with age-matched controls and analyze data in a sex-stratified manner to account for these variables.

Sample Size Calculation: Base power analyses on reported precision metrics for GLRX assays. For ELISA methods, intra-assay precision (CV<8%) and inter-assay precision (CV<10%) provide a foundation for calculating required sample sizes .

Intervention Timeline: GLRX regulation involves both rapid post-translational modifications (minutes to hours) and longer-term transcriptional responses (hours to days). Design sampling timepoints accordingly, with more frequent early sampling to capture rapid changes.

Multiple Readouts: Combine protein quantification (Western blot/ELISA) with activity assays and transcriptional analysis (qPCR) to capture the full spectrum of GLRX regulation.

Environmental Factors: Control for environmental variables that affect redox status, including diet, housing conditions, and exposure to environmental stressors.

How can I differentiate between changes in GLRX expression versus activity?

Distinguishing between alterations in GLRX expression versus activity requires parallel analytical approaches:

Protein Quantification: Western blot and ELISA measure GLRX protein levels but provide no information about enzymatic function. Specific antibodies can detect GLRX at approximately 12 kDa in multiple mouse cell lines including NIH-3T3 and C2C12 .

Activity Assays: Standard GLRX activity assays measure the rate of substrate reduction in the presence of GSH, NADPH, and glutathione reductase. These provide direct measurement of catalytic function regardless of protein level.

Correlation Analysis: Calculate the ratio of activity to protein concentration (specific activity) across experimental groups. Changes in this ratio suggest post-translational modifications affecting enzyme function.

Redox State Assessment: Analyze the redox state of GLRX itself through non-reducing gels or redox proteomics approaches. Oxidative modifications can inhibit GLRX activity without changing protein levels.

Subcellular Localization: Changes in GLRX distribution between cellular compartments can alter local activity without affecting total protein levels. Subcellular fractionation followed by compartment-specific analysis can reveal such changes.

mRNA-Protein Correlation: Compare changes in GLRX mRNA levels with protein levels. Discordance suggests post-transcriptional regulation mechanisms.

How does GLRX function in mouse models of infection and inflammation?

GLRX plays complex roles in mouse models of infection and inflammation:

Species-Specific Inflammatory Responses: Comparative studies between mice, rats, and deermice reveal important species differences in inflammatory responses. While laboratory mice and rats show activation of endogenous retroviruses (ERVs) after LPS exposure, white-footed deermice (Peromyscus leucopus) do not exhibit this response . This species difference may explain the ability of deermice to tolerate infections that cause pathology in other species.

Regulatory Role in NF-κB Signaling: In mouse models, GLRX regulates the NF-κB pathway through deglutathionylation of key components, influencing inflammatory gene expression. This becomes particularly important during bacterial infections when TLR4 signaling (activated by LPS) triggers both NF-κB activation and oxidative stress.

Temporal Dynamics: GLRX exhibits distinct temporal patterns during inflammation in mice. Initial oxidative bursts may temporarily decrease GLRX activity through oxidative modifications, while prolonged inflammation typically induces compensatory increases in GLRX expression as part of the antioxidant response.

Cell-Type Specific Functions: In immune cells, GLRX regulates redox-sensitive transcription factors differently in various subpopulations. For instance, GLRX expression in mouse macrophages affects their polarization toward pro-inflammatory or anti-inflammatory phenotypes during infection.

Tissue-Specific Protection: In mouse lung inflammation models, GLRX helps protect epithelial cells against oxidative damage, while in vascular inflammation, it modulates endothelial activation and leukocyte adhesion through redox regulation of adhesion molecules.

What challenges exist in comparing mouse and human GLRX systems?

Comparing mouse and human GLRX systems presents several methodological challenges:

Antibody Cross-Reactivity Considerations: Some antibodies detect both human and mouse GLRX1, as seen with reagents that recognize recombinant human GLRX1 spanning Met1-Gly106 . While useful for comparative studies, this cross-reactivity requires careful validation to ensure specificity.

Expression Pattern Differences: The tissue distribution pattern of GLRX differs between mice and humans. These differences must be considered when translating findings from mouse models to human pathologies.

Functional Conservation Assessment: Despite structural similarities, subtle differences in substrate specificity exist between mouse and human GLRX. Comparative functional assays using identical substrates are necessary to establish true functional equivalence.

Cross-Species Sample Analysis: When analyzing mixed species samples, technical challenges can arise. In gene expression studies, human-mouse mixed samples show bimodal distribution patterns that require specialized analytical approaches . Similar considerations may apply to protein-level studies.

Intervention Response Variations: Pharmacological agents targeting redox systems may have different potency, specificity, or pharmacokinetics between species, complicating translational research.

How can single-cell approaches advance our understanding of GLRX in mouse models?

Single-cell approaches offer powerful new insights into GLRX biology:

Heterogeneity Revelation: Single-cell RNA sequencing can reveal cell-specific GLRX expression patterns within heterogeneous tissues. This is particularly valuable for tissues with complex cellular composition, where bulk analysis obscures important cell-type-specific regulation.

Correlation with Cell State Markers: Single-cell approaches allow correlation of GLRX expression with markers of cell activation, stress response, or differentiation state at the individual cell level, revealing conditional regulation patterns.

Spatial Context Integration: Combining single-cell transcriptomics with spatial techniques (spatial transcriptomics or immunohistochemistry) provides insight into how microenvironmental factors influence GLRX expression within tissues.

Trajectory Analysis: Single-cell trajectory analysis can reveal dynamic changes in GLRX expression during cell state transitions, such as during immune cell activation or differentiation processes.

Cross-Species Analytical Challenges: When analyzing mixed human-mouse samples, specialized pipelines are needed to correctly classify cells and account for probe cross-reactivity. As documented in methodological studies, "true organism probes will have much higher affinity for the transcripts than the cross-hybridized ones" .

Why might I see inconsistent GLRX detection in my mouse samples?

Inconsistent GLRX detection can stem from several factors:

Sample Processing Variability: GLRX is sensitive to oxidation during sample preparation. Inconsistent use of reducing agents or delays in processing can cause variable detection. Standardize processing times and maintain consistent temperature throughout.

Antibody Selection Issues: For Western blotting, verify specificity using recombinant protein standards. Validated antibodies have demonstrated consistent detection of GLRX at approximately 12 kDa in multiple mouse cell lines including NIH-3T3 mouse embryonic fibroblasts and C2C12 mouse myoblasts .

Post-translational Modifications: GLRX undergoes various modifications (glutathionylation, oxidation) that can affect antibody recognition. These modifications may vary with physiological state and sample handling.

Matrix Effects in ELISA: Different sample matrices show variable recovery rates:

  • Serum: 79-93% recovery (average 86%)

  • EDTA plasma: 90-105% recovery (average 97%)

  • Heparin plasma: 86-99% recovery (average 92%)

These differences necessitate matrix-specific calibration and consistent sample preparation.

Detection Range Limitations: For ELISA methods, samples must fall within the assay's linear range (typically 78.13-5000 pg/mL) . Concentrations outside this range require appropriate dilution.

Storage Degradation: Multiple freeze-thaw cycles significantly degrade GLRX. Aliquot samples upon initial processing to avoid repeated freezing and thawing.

What statistical approaches are recommended for analyzing GLRX data across experimental groups?

Appropriate statistical analysis of mouse GLRX data requires consideration of several factors:

Normality Testing: Begin with tests for normality (Shapiro-Wilk or Kolmogorov-Smirnov) to determine whether parametric or non-parametric approaches are appropriate. GLRX expression data often follow log-normal distributions.

Multiple Group Comparisons: For comparing GLRX across experimental groups:

  • For normally distributed data: One-way ANOVA followed by appropriate post-hoc tests

  • For non-parametric data: Kruskal-Wallis followed by Dunn's test

Time-Course Analysis: For experiments measuring GLRX at multiple timepoints, use repeated measures ANOVA or mixed-effects models that account for within-subject correlations.

Precision Considerations: Base power calculations on documented precision metrics:

  • Intra-assay precision: CV<8%

  • Inter-assay precision: CV<10%

These values help determine the minimum sample size needed to detect biologically meaningful differences.

Correlation Analysis: When examining relationships between GLRX and other parameters (oxidative stress markers, inflammatory cytokines), use appropriate correlation tests based on data distribution.

Multiple Testing Correction: When analyzing GLRX across multiple tissues or conditions, apply correction for multiple comparisons (Benjamini-Hochberg procedure) to control false discovery rate.

How should I interpret GLRX changes in the context of inflammation and infection models?

Interpreting GLRX changes during inflammation requires contextual analysis:

Species-Specific Response Patterns: Consider that different rodent species show distinctive inflammatory response patterns. While mice and rats show significant upregulation of endogenous retroviruses (ERVs) after LPS exposure, white-footed deermice (Peromyscus leucopus) do not exhibit this response . These differences in inflammatory pathway activation likely affect GLRX regulation as well.

Temporal Context: Early GLRX changes (0-6 hours post-stimulus) often reflect post-translational regulation, while later changes (12-24+ hours) typically indicate transcriptional regulation. Compare your timepoints to this framework.

Pathway Integration: Interpret GLRX changes alongside other inflammatory markers (cytokines, acute phase proteins) and redox systems (GSH/GSSG ratio, thioredoxin system). This provides context for whether GLRX changes are driving or responding to inflammation.

Tissue-Specific Interpretation: The same absolute change in GLRX may have different functional implications in different tissues based on baseline expression levels and the tissue's metabolic characteristics.

Treatment Response Assessment: In intervention studies, determine whether GLRX changes precede, coincide with, or follow changes in clinical parameters or other molecular markers. This temporal relationship provides insight into whether GLRX modulation is causative or responsive.

Cross-Species Translation: When using mouse models to understand human disease, consider that inflammatory responses show species-specific patterns . Validate key findings across multiple species when possible to strengthen translational relevance.

What are the key considerations for designing robust GLRX studies in mouse models?

Designing robust GLRX studies in mouse models requires integrated consideration of technical and biological factors. Methodologically, researchers must select appropriate detection techniques based on their specific research questions, with Western blot providing qualitative assessment, ELISA offering quantitative measurement within specific sensitivity ranges (35-5000 pg/mL), and activity assays delivering functional insights.

Sample preparation represents a critical variable, with tissue-specific extraction protocols, appropriate dilution factors, and preservation of native redox state all essential for reliable results. Experimental design must account for mouse strain differences, age and sex effects, and appropriate controls including vehicle treatments and time-matched sampling.

The complex regulation of GLRX during inflammation and infection necessitates comprehensive analysis of both expression and activity, ideally across multiple timepoints to capture dynamic responses. Finally, interpreting GLRX changes requires contextualizing them within broader inflammatory and redox networks, with species differences always considered when translating findings to human biology.

By addressing these considerations systematically, researchers can generate more reproducible and translatable insights into the role of GLRX in health and disease.

What emerging approaches might advance mouse GLRX research in the near future?

Several emerging approaches promise to advance mouse GLRX research:

  • Single-cell resolution techniques will increasingly reveal cell-type-specific GLRX regulation that is obscured in bulk tissue analysis. Specialized protocols for fixed and frozen samples, like the 10x Genomics Gene Expression Flex kit, are making these approaches more accessible .

  • CRISPR-based in vivo editing will enable more precise manipulation of GLRX and related pathways in mouse models, moving beyond traditional knockout approaches to study specific regulatory elements or post-translational modifications.

  • Integration of multi-omics data (transcriptomics, proteomics, metabolomics) will provide comprehensive views of how GLRX functions within broader redox networks during inflammation, infection, and other stress responses.

  • Comparative immunology approaches examining inflammatory differences between species, as highlighted in studies comparing laboratory mice with naturally disease-tolerant species like white-footed deermice , may reveal evolutionary insights into redox regulation during infection.

  • Development of redox-sensitive biosensors for real-time imaging of GLRX activity in living tissues will transform our understanding of dynamic redox regulation in various physiological and pathological contexts.

Product Science Overview

Introduction

Glutaredoxins (GRXs) are small, ubiquitous proteins that play a crucial role in maintaining cellular redox homeostasis. They are part of the thioredoxin superfamily and are involved in various cellular processes, including DNA synthesis, signal transduction, and defense against oxidative stress. This article focuses on the background of mouse recombinant glutaredoxin, highlighting its structure, function, and significance in biological research.

Structure and Classification

Glutaredoxins are characterized by their conserved active site motif, typically CXXC or CXXS, where “C” stands for cysteine and “X” can be any amino acid. Based on their active sites, GRXs are classified into three subfamilies: CPYC, CGFS, and CC . The CPYC and CGFS subfamilies are present in eukaryotes, while the CC subfamily is exclusive to higher plants .

Expression and Purification

Recombinant mouse glutaredoxin is commonly expressed in Escherichia coli (E. coli) systems due to the ease of genetic manipulation and high yield of protein production . The recombinant protein is typically purified using affinity chromatography techniques, ensuring high purity and biological activity .

Biological Functions

Glutaredoxins are primarily involved in the reduction of disulfide bonds in proteins, utilizing glutathione (GSH) as a cofactor. They play a pivotal role in:

  • Redox Homeostasis: Maintaining the balance between oxidation and reduction within the cell.
  • DNA Synthesis: Supporting ribonucleotide reductase activity, which is essential for DNA synthesis .
  • Signal Transduction: Modulating various signaling pathways by regulating the redox state of target proteins.
  • Oxidative Stress Response: Protecting cells from oxidative damage by reducing reactive oxygen species (ROS) and repairing oxidized proteins .
Research Significance

Recombinant mouse glutaredoxin is widely used in research to study its role in cellular processes and its potential therapeutic applications. It serves as a model to understand the function of GRXs in higher organisms and their involvement in diseases such as cancer and neurodegenerative disorders .

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.