GSTM1 Mouse

Glutathione S-Transferase M1 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to GSTM1 Mouse

GSTM1 (Glutathione S-Transferase Mu 1) Mouse refers to the murine isoform of the GST enzyme critical for detoxifying electrophilic compounds, including environmental toxins, carcinogens, and oxidative stress byproducts. This enzyme belongs to the mu class of GSTs, which are evolutionarily conserved and highly expressed in mouse kidneys and liver . Recombinant GSTM1 Mouse proteins and knockout (KO) models are widely used to study oxidative stress-related diseases, pharmacokinetics, and metabolic pathways .

Protein Structure

  • Source: Recombinant GSTM1 Mouse is produced in E. coli as a single, non-glycosylated polypeptide chain containing 218 amino acids (25.9 kDa) or 238 amino acids (28.1 kDa) with an N-terminal His-Tag .

  • Sequence: Includes conserved residues critical for glutathione binding and catalytic activity (e.g., MPMILGYWNV...SNK ).

Biochemical Properties

PropertyValue
Molecular Mass25.9 kDa (native), 28.1 kDa (His-Tag)
Purity>95% (SDS-PAGE)
Specific Activity3 units/mg (CDNB conjugation)
Storage4°C (short-term), -20°C (long-term)
Endotoxin Levels<1.0 EU/μg

Detoxification and Oxidative Stress

GSTM1 conjugates glutathione to reactive oxygen species (ROS) and lipid peroxidation products like 4-hydroxy-2-nonenal, mitigating oxidative damage . Its absence (Gstm1-KO) leads to:

  • Elevated renal superoxide levels .

  • Increased susceptibility to angiotensin II-induced hypertension and cisplatin-induced acute kidney injury (AKI) .

  • Exaggerated glomerulosclerosis and tubulointerstitial inflammation in chronic kidney disease (CKD) models .

Role in Disease Models

  • Hypertension: Gstm1-KO mice exhibit worsened kidney injury and inflammation independent of blood pressure .

  • Obesity: GSTM1 knockdown promotes white adipose tissue (WAT) browning, reducing adipogenesis and improving metabolic health .

  • Neuroinflammation: GSTM1 modulates TNF-α-dependent astrocyte activation, influencing neuronal stress and cognitive decline .

Phenotypic Comparison: Wild-Type vs. Gstm1-KO Mice

ParameterWild-Type MiceGstm1-KO Mice
Renal Oxidative StressBaseline levels2–3x increase
Survival Post-CKD100% at 13 weeks<40% at 8 weeks
Glutathione ActivityNormal GST-D activityNear-absent in liver/kidney
Response to SulforaphaneMild renal protectionSignificant protection

Mechanistic Insights

  • CKD Pathogenesis: Gstm1 deficiency increases ROS-driven renal inflammation (e.g., elevated MCP-1, IL-6, and neutrophil infiltration) .

  • Drug Toxicity: Gstm1/Gstt1 double-KO mice show heightened sensitivity to cisplatin nephrotoxicity due to impaired detoxification .

  • Gene Regulation: GSTM1 knockdown in astrocytes alters TNF-α-induced transcriptional programs, downregulating pro-inflammatory genes (e.g., CXCL1, CSF2) .

Applications of Recombinant GSTM1 Mouse Protein

  • Enzyme Activity Assays: Used to quantify GST activity via CDNB conjugation .

  • Drug Metabolism Studies: Evaluates detoxification pathways for xenobiotics and chemotherapeutics .

  • Therapeutic Development: Screens compounds targeting oxidative stress (e.g., sulforaphane) .

Product Specs

Introduction
The soluble cytoplasmic mammalian GSTs are divided into eight classes: alpha, kappa, mu, omega, pi, sigma, theta, and zeta. Two different supergene families encode cytosolic and membrane-bound GST types. The detoxification of electrophilic compounds, such as carcinogens, therapeutic medications, environmental toxins, and oxidative stress products, by conjugation with glutathione, is carried out by the mu class of enzymes. On chromosome 1p13.3, a gene cluster containing the genes that code for the mu class of enzymes is highly polymorphic. These genetic variations may affect an individual's susceptibility to toxins and carcinogens and the effectiveness and toxicity of some medications. The development of several cancers has been linked to null mutations in this mu class gene.
Description
GSTM1 Mouse Recombinant is a single, non-glycosylated polypeptide chain with a molecular weight of 25.9 kDa that contains 218 amino acids. It is produced in E. coli. Proprietary chromatographic methods are used to purify the GTM1.
Physical Appearance
Colorless solution that has been sterile filtered.
Formulation
PBS pH-7.4 and 5mM glutathione are present in the GSTM1 solution.
Stability
Store at 4°C for 2-4 weeks if the entire vial will be used. It is advised to add a carrier protein (0.1% HSA or BSA) for long-term storage. For extended storage, keep frozen at -20°C. Avoid repeated freeze-thaw cycles.
Purity
Greater than 95.0% as determined by SDS-PAGE analysis.
Biological Activity
The specific activity, which is measured at pH 6.5 and 25°C, is 3 units/mg. It is defined as the quantity of enzyme that conjugates 1.0 umole of 1-chloro-2,4-dinitrobenzene (CDNB) with reduced glutathione per minute.
Synonyms
GST1, GTH4, GTM1, GSTM1-1, MGC26563, GSTM1a-1a, GSTM1b-1b, GSTM1, Glutathione S-transferase Mu 1, GST class-mu 1, Glutathione S-transferase GT8.7, pmGT10, GST 1-1.
Source
Escherichia Coli.
Amino Acid Sequence
MPMILGYWNV RGLTHPIRML LEYTDSSYDE KRYTMGDAPD FDRSQWLNEK FKLGLDFPNL PYLIDGSHKI TQSNAILRYL ARKHHLDGET EEERIRADIV ENQVMDTRMQ LIMLCYNPDF EKQKPEFLKT IPEKMKLYSE FLGKRPWFAG DKVTYVDFLA YDILDQYRMF EPKCLDAFPN LRDFLARFEG LKKISAYMKS SRYIATPIFS KMAHWSNK.

Q&A

What is GSTM1 and what are its primary functions in mice?

GSTM1 (Glutathione S-Transferase Mu 1) is an enzyme belonging to the GST family that plays a critical role in phase II detoxification reactions. In mice, GSTM1 shares approximately 80% amino acid identity with human GSTM1 and >90% with rat GSTM1 . This enzyme catalyzes the conjugation of reduced glutathione to various substrates, including xenobiotics and products of oxidative stress, facilitating their elimination from the body. GSTM1 is highly expressed in mouse liver and kidney, where it serves as a key enzyme for phase II biotransformation reactions . The primary functions of GSTM1 include detoxification of environmental toxins, protection against oxidative stress, and maintenance of cellular redox homeostasis. Under normal physiological conditions, GSTM1 contributes to the body's antioxidant defense system by neutralizing reactive oxygen species and preventing oxidative damage to cellular components .

How are GSTM1 knockout mouse models generated?

GSTM1 knockout (KO) mouse models can be generated through several approaches, with gene targeting being the most common method. According to the literature, one approach involves the electroporation of embryonic stem cells derived from specific mouse strains (e.g., 129/SvEv) with a targeting construct designed to disrupt the Gstm1 locus . The targeting construct typically disrupts a segment of the Gstm1 gene, rendering it non-functional. After electroporation, successfully targeted stem cells are identified and injected into blastocysts to generate chimeric mice. These chimeras are then bred to establish germline transmission of the mutant allele. Confirmation of Gstm1 ablation is typically performed using Southern blotting and quantitative RT-PCR techniques to verify the absence of Gstm1 mRNA expression . Modern CRISPR/Cas9 approaches may also be employed for more targeted and efficient gene disruption, although the traditional embryonic stem cell approach remains well-documented in the literature for Gstm1 knockout generation.

What phenotypic changes are observed in GSTM1 knockout mice under baseline conditions?

Under baseline conditions, GSTM1 knockout mice exhibit several distinct phenotypic changes compared to wild-type controls. These mice generally appear normal and remain fertile , but display subtle physiological differences. Most notably, GSTM1 KO mice show a modest but statistically significant elevation in systolic blood pressure (approximately 7 mm Hg higher than wild-type controls) . They also demonstrate increased oxidative stress, as evidenced by higher urinary 8-isoprostane levels, a recognized marker of oxidative stress .

How is GSTM1 protein expression typically measured in mouse tissues?

GSTM1 protein expression in mouse tissues can be quantified using several complementary techniques. Enzyme-linked immunosorbent assay (ELISA) is a commonly employed method, with commercial kits available that can detect mouse GSTM1 in various sample types including serum, plasma, tissue homogenates, cell lysates, and cell culture supernatants . These sandwich immunoassays typically have high sensitivity (as low as 0.57 ng/mL) and a detection range of 1.57-100 ng/mL .

Western blotting represents another standard approach for GSTM1 protein detection, using specific antibodies to visualize and quantify GSTM1 expression in tissue lysates. Immunohistochemistry or immunofluorescence staining can be used to determine the tissue and cellular distribution patterns of GSTM1. For functional assessment, GSTM1 enzyme activity can be measured using specific substrates such as DCNB or CDNB in cytosolic fractions prepared from mouse tissues. The activity toward these substrates is significantly reduced in GSTM1 knockout mice, providing a functional readout of GSTM1 expression . Additionally, mass spectrometry-based proteomic approaches offer the advantage of absolute quantification and can be particularly useful for detecting potential compensatory changes in other GST family members.

What are the key methodological considerations when studying GSTM1 in mouse models of disease?

When studying GSTM1 in mouse models of disease, several methodological considerations are critical for robust experimental design and data interpretation. First, researchers should carefully select appropriate disease models based on the known functions of GSTM1. Since GSTM1 is involved in detoxification and antioxidant defense, models featuring oxidative stress components (such as chronic kidney disease, cardiovascular disease, or toxicant exposure) are particularly relevant .

Sex-specific effects must be considered, as the literature indicates that mechanisms compensating for GSTM1 loss differ between male and female mice . Experiments should include both sexes or provide appropriate justification for sex selection. The genetic background of mouse models is another crucial factor, as it can significantly influence the phenotypic manifestation of GSTM1 deletion. Researchers should maintain consistent genetic backgrounds or use appropriate backcrossing strategies.

Tissue-specific effects should be considered when designing experiments and interpreting results. The literature suggests that GSTM1 deletion has different consequences in different tissues . Therefore, multi-tissue analyses may be necessary for comprehensive understanding. Additionally, age-dependent effects should be accounted for, as some phenotypes may only become apparent with aging. For instance, at one year of age, no differences in mortality or phenotypes were observed between wild-type and GSTM1 knockout mice under baseline conditions .

Finally, appropriate controls must be included, such as wild-type littermates and, when applicable, heterozygous mice to assess gene dosage effects. Researchers should also consider measuring compensatory changes in other GST family members, as these may confound interpretation of GSTM1-specific effects.

How does GSTM1 deletion affect susceptibility to specific disease models in mice?

GSTM1 deletion significantly increases susceptibility to disease progression in several mouse models, with particularly pronounced effects in models involving oxidative stress and inflammation. In the context of chronic kidney disease (CKD), GSTM1 knockout mice subjected to 5/6 nephrectomy (Nx-CKD model) exhibit dramatically worse outcomes compared to wild-type controls. These mice show significantly reduced survival rates, with only 23% surviving to 83 days post-procedure compared to 100% survival in wild-type mice at 90 days . This increased mortality is accompanied by exacerbated hypertension and more severe kidney injury.

In cardiovascular models, the effects of GSTM1 deletion appear to be sex-dependent. In a Langendorff-perfused cardiac ischemia-reperfusion injury model, male GSTM1 knockout hearts display unexpected protection against injury, while female knockout hearts show no such protection . This sexual dimorphism in cardiovascular response highlights the complex role of GSTM1 in tissue-specific redox regulation and suggests distinct compensatory mechanisms between sexes.

GSTM1 also influences neurological outcomes in models of neuroinflammation. Knockdown of GSTM1 in astrocytes increases neuronal stress levels and attenuates neuronal activities during lipopolysaccharide (LPS)-induced inflammation . This suggests a neuroprotective role for GSTM1 in the context of inflammatory challenges to the central nervous system.

Additionally, GSTM1 appears to be essential for reducing cisplatin-induced ototoxicity in CBA/CaJ mice, indicating its importance in protecting against chemotherapy-induced hearing loss . This protection likely relates to GSTM1's role in detoxifying reactive intermediates generated during cisplatin metabolism.

What metabolic and transcriptional changes occur in GSTM1 knockout mice?

GSTM1 knockout mice exhibit distinct metabolic and transcriptional signatures that vary by tissue type and sex. Metabolomic analyses have revealed tissue-specific alterations in various metabolic pathways. In hearts from male GSTM1 knockout mice, increases in several metabolites with antioxidant properties have been observed, including carnosine and anserine. These changes were not observed in female hearts, highlighting sex-specific metabolic adaptations .

In the context of hypertension, GSTM1 knockout mice show significant increases in metabolites involved in pathways linking methionine, cysteine, and glutathione metabolism in the kidney. These changes were not observed in heart tissue, demonstrating tissue-specific metabolic responses to GSTM1 deletion . After administration of xenobiotics like 1,2-dichloro-4-nitrobenzene (DCNB), GSTM1 null mice show altered pharmacokinetics with larger plasma AUC0-24 (5.1-5.3 times that of wild-type controls) and higher Cmax (2.1-2.2 times wild-type levels), with correspondingly lower levels of glutathione-related metabolites .

At the transcriptional level, RNA-seq analysis of cultured astrocytes with reduced GSTM1 expression has revealed altered gene expression patterns, particularly in response to inflammatory stimuli like TNF-α. GSTM1 knockdown astrocytes show a larger number of differentially expressed genes following TNF-α stimulation compared to control cells . Gene set enrichment analysis of these differentially expressed genes points to alterations in multiple pathways related to inflammation and cellular stress responses. Additionally, a small decrease in glutathione S-transferase alpha 3 mRNA expression has been noted in GSTM1 null mice, suggesting modest compensatory regulation among GST family members .

How do compensatory mechanisms mitigate the effects of GSTM1 deletion in different tissues and sexes?

Compensatory mechanisms following GSTM1 deletion exhibit remarkable tissue and sex specificity, highlighting the complex redundancy within the glutathione S-transferase system. In male hearts, but not female hearts, GSTM1 knockout leads to upregulation of other GST family members as detected by qPCR analysis . This transcriptional compensation likely contributes to the unexpected cardioprotection observed in male GSTM1 knockout mice subjected to ischemia-reperfusion injury. In contrast, such compensatory upregulation of other GST genes is not observed in male kidney tissue, potentially explaining the increased susceptibility of GSTM1 knockout mice to kidney injury .

Metabolomic analyses further illuminate these tissue-specific compensatory mechanisms. Male GSTM1 knockout hearts show increases in endogenous antioxidant metabolites, particularly carnosine and anserine, which are not observed in female hearts . These metabolites may provide alternative antioxidant protection in the absence of GSTM1. Other antioxidant-related metabolites show increased levels in hearts of both sexes but not in kidneys, again emphasizing tissue-specific metabolic adaptation.

In the context of hypertension, GSTM1 knockout kidneys display significant alterations in metabolites involved in pathways connecting methionine, cysteine, and glutathione metabolism . These changes suggest engagement of alternative detoxification pathways to compensate for GSTM1 deficiency under stress conditions. The molecular basis for these sex and tissue-specific differences in compensatory response remains incompletely understood but likely involves differential regulation of transcription factors controlling antioxidant response elements, such as Nrf2, which has been shown to be upregulated along with GSTM1 in wild-type mice following 5/6 nephrectomy .

What are the challenges in translating findings from GSTM1 knockout mice to human GSTM1 polymorphisms?

Translating findings from GSTM1 knockout mice to human GSTM1 polymorphisms presents several significant challenges that researchers must carefully address. First, there are important structural and functional differences between mouse and human GSTM1 proteins. The murine GSTM1 shares approximately 80% amino acid identity with human GSTM1 , suggesting potentially important differences in substrate specificity and catalytic efficiency. These molecular differences may result in distinct physiological roles and responses to stressors between species.

The prevalence of GSTM1 deletion also varies substantially between human populations, whereas experimental mouse models are typically studied on defined genetic backgrounds. Population stratification and diverse genetic architectures in humans might influence how GSTM1 deletion manifests phenotypically across different ethnic groups. Furthermore, complex gene-environment interactions likely modify the effects of GSTM1 deletion in humans in ways that are difficult to model in laboratory mice. For instance, the impact of GSTM1 polymorphisms may depend on dietary factors such as cruciferous vegetable intake, which provides compounds that interact with glutathione S-transferase pathways .

Finally, differences in disease pathophysiology between mice and humans can complicate translation. While mouse models provide valuable insights into mechanisms, the progression and manifestation of complex diseases often differ between species, potentially altering the specific contribution of GSTM1 deficiency to disease phenotypes.

How does GSTM1 interact with inflammatory pathways in different experimental models?

GSTM1 demonstrates complex interactions with inflammatory pathways across various experimental models, revealing its multifaceted role beyond simple detoxification. In neuroinflammatory models, GSTM1 appears to modulate astrocyte activation and subsequent neuronal function. RNA-sequencing analysis of astrocytes with GSTM1 knockdown revealed altered transcriptional responses to TNF-α stimulation compared to control cells . GSTM1-deficient astrocytes exhibited changes in genes involved in inflammatory signaling, with consequences for neuronal stress and activity. Specifically, GSTM1 reduction in astrocytes increased neuronal stress levels and attenuated neuronal activities during lipopolysaccharide (LPS)-induced inflammation .

In kidney disease models, GSTM1 knockout mice subjected to 5/6 nephrectomy display exacerbated inflammatory responses alongside increased oxidative stress. This suggests that GSTM1 normally functions to suppress excessive inflammation following kidney injury, potentially through mechanisms involving redox-sensitive inflammatory signaling pathways .

The relationship between GSTM1 and inflammation appears bidirectional. While GSTM1 deficiency can promote inflammation, inflammatory stimuli may also regulate GSTM1 expression. This creates potential feedback loops in disease progression. The mechanisms linking GSTM1 function to inflammatory signaling likely involve its influence on cellular redox status, as many inflammatory transcription factors and signaling molecules (such as NF-κB and MAP kinases) are redox-sensitive.

Interestingly, GSTM1's impact on inflammation exhibits tissue and sex specificity. The differential effects observed in male versus female tissues following GSTM1 deletion suggest that hormonal or other sex-specific factors may modify how GSTM1 deficiency influences inflammatory cascades. These complex interactions highlight the importance of considering GSTM1 not merely as a detoxification enzyme but as an integrated component of cellular stress response and inflammatory signaling networks.

What methodological approaches can detect subtle phenotypes in GSTM1 knockout mice?

Detecting subtle phenotypes in GSTM1 knockout mice requires sophisticated methodological approaches that extend beyond standard observational techniques. Continuous physiological monitoring systems, such as radiotelemetry for blood pressure measurement, can reveal modest but significant differences that might be missed with intermittent measurements. For example, radiotelemetry detected a 7 mm Hg elevation in systolic blood pressure in GSTM1 knockout mice compared to wild-type controls , a difference that could be overlooked with less sensitive methods.

Stress challenge protocols represent another valuable approach for unmasking latent phenotypes. While GSTM1 knockout mice may appear normal under baseline conditions, challenging these mice with stressors like disease models (e.g., 5/6 nephrectomy) , xenobiotic administration , or inflammatory stimuli can reveal significant vulnerabilities. These challenge protocols often unmask phenotypes not apparent under standard housing conditions.

Sensitive biochemical assays for oxidative stress markers provide important insights into the molecular consequences of GSTM1 deletion. Measurements of 8-isoprostane levels in urine , 4-hydroxynonenal (4-HNE) immunostaining , or advanced metabolomic profiling can detect alterations in redox status that precede overt pathological changes. Multi-omics approaches, including transcriptomics, proteomics, and metabolomics, offer comprehensive views of molecular changes resulting from GSTM1 deletion. These approaches have revealed tissue and sex-specific alterations in gene expression and metabolite profiles that help explain phenotypic differences .

Age-dependent phenotyping is crucial, as some effects of GSTM1 deletion may only become apparent with aging or cumulative exposure to environmental stressors. Finally, sophisticated imaging techniques, such as intravital multiphoton microscopy for kidney studies or advanced neuroimaging, can visualize functional and structural changes at cellular and tissue levels that might be missed with conventional histology.

What are the current technical limitations in studying GSTM1 function in mice?

Despite significant advances in mouse genetic models, several technical limitations persist in studying GSTM1 function. The functional redundancy within the GST family represents a major challenge, as other GST isoforms may compensate for GSTM1 deletion, potentially masking phenotypes . This redundancy necessitates sophisticated approaches to disentangle the specific contributions of GSTM1 from compensatory mechanisms involving other GST family members.

The tissue-specific and cell type-specific expression patterns of GSTM1 complicate the interpretation of whole-animal knockout models. Global GSTM1 knockout mice may exhibit complex phenotypes reflecting the combined effects of GSTM1 deficiency across multiple tissues, making it difficult to isolate tissue-specific contributions. While conditional knockout models could address this limitation, such models for GSTM1 have not been extensively reported in the literature.

Temporal aspects of GSTM1 function present additional challenges. Constitutive knockout models cannot distinguish between developmental effects and adult functions of GSTM1. Inducible knockout systems would be valuable but are not widely reported for GSTM1 studies. The high variability in environmental exposure to GSTM1 substrates across experimental settings may contribute to inconsistent phenotypes between studies. Standardized environmental conditions and exposure protocols would improve reproducibility.

Limitations in detecting and quantifying GSTM1 protein also exist. While antibody-based methods like ELISA provide quantification , cross-reactivity with other GST family members remains a concern due to sequence homology. The lack of highly specific activity assays for GSTM1 further complicates functional assessment. Current enzymatic assays with substrates like DCNB and CDNB are not completely specific to GSTM1 and may reflect activities of multiple GST isoforms.

Finally, translating findings from mouse models to human disease is complicated by species differences in GSTM1 structure, expression patterns, and substrate preferences, as well as differences in the genetic context of GSTM1 deletion between engineered mouse models and human polymorphisms .

Product Science Overview

Introduction

Glutathione S-Transferase M1 (GSTM1) is a member of the glutathione S-transferase (GST) family, which plays a crucial role in detoxification processes. GSTs are enzymes that catalyze the conjugation of the antioxidant molecule glutathione (GSH) to various endogenous and exogenous electrophilic compounds. This process is essential for the detoxification of harmful substances and the protection of cellular components from oxidative damage.

Structure and Function

GSTM1 is a phase II detoxification enzyme predominantly expressed in the liver. It is involved in the metabolism of a wide range of substrates, including carcinogens, therapeutic drugs, environmental toxins, and products of oxidative stress. The enzyme’s activity is crucial for maintaining cellular homeostasis and protecting against cellular damage.

The structure of GSTM1 includes a highly conserved GSH-binding site and a more variable substrate-binding site. This dual binding capability allows GSTM1 to interact with a diverse array of substrates, making it a versatile enzyme in detoxification pathways.

Recombinant Expression

Recombinant GSTM1 (Mouse) is produced using Escherichia coli expression systems. The recombinant protein is typically purified to a high degree of purity (>95%) and is suitable for various biochemical assays, including SDS-PAGE. The availability of recombinant GSTM1 allows for detailed studies on its enzymatic properties, substrate specificity, and potential applications in biotechnology and medicine .

Biological Significance

GSTM1 plays a significant role in protecting cells from oxidative stress and toxic insults. It is involved in the detoxification of reactive oxygen species (ROS) and the metabolism of xenobiotics. The enzyme’s activity is particularly important in the liver, where it helps to neutralize harmful compounds and prevent liver damage.

Research has shown that GSTM1 is upregulated in response to chronic alcohol consumption, providing a protective mechanism against alcohol-induced liver injury. The enzyme counteracts the activation of apoptosis signal-regulating kinase 1 (ASK1) and its downstream signaling pathways, thereby mitigating oxidative stress and inflammation in the liver .

Applications in Research and Medicine

Recombinant GSTM1 is widely used in research to study its role in detoxification processes and its potential therapeutic applications. The enzyme’s ability to neutralize harmful compounds makes it a valuable tool in the development of treatments for diseases associated with oxidative stress and toxin exposure.

In addition, GSTM1 has potential applications in biotechnology, such as the development of biosensors for detecting environmental toxins and the engineering of plants with enhanced resistance to biotic and abiotic stresses .

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.