GSTM1 Mouse, His

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

Description

Detoxification and Oxidative Stress

GSTM1 catalyzes glutathione conjugation to neutralize electrophilic toxins, including reactive oxygen species (ROS) and lipid peroxidation byproducts like 4-hydroxy-2-nonenal (4-HNE) . Key functions include:

  • Xenobiotic Metabolism: Detoxifies environmental carcinogens and therapeutic drugs .

  • Redox Homeostasis: Mitigates oxidative stress in tissues such as kidneys and brain .

Disease Associations

ConditionRole of GSTM1 DeficiencyEvidence
Chronic Kidney Disease (CKD)Accelerated glomerulosclerosis, inflammation, and mortality in Gstm1 knockout (KO) mice .KO mice showed 60% mortality by 8 weeks post-nephrectomy vs. 0% in wild-type .
HypertensionElevated systolic blood pressure (+7 mmHg) and urinary 8-isoprostane (oxidative stress marker) .Linked to impaired vascular smooth muscle cell regulation .
NeuroinflammationReduced neuronal activity (c-Fos expression) and increased lipid peroxidation (4-HNE) in astrocytes .GSTM1 knockdown in astrocytes altered TNF-α-mediated gene networks .

Experimental Models

  • Kidney Injury Studies: Gstm1 KO mice exhibit exacerbated renal damage in ischemia-reperfusion and angiotensin II-induced hypertension models .

  • Neuroinflammation: Astrocyte-specific GSTM1 knockdown models reveal its role in modulating neuronal stress and interferon responses .

  • Aging: Increased GSTM1 expression in aging mouse brains correlates with memory decline .

Key Findings

  • Oxidative Stress: GSTM1 deficiency elevates ROS and lipid peroxidation adducts, worsening tissue injury .

  • Gene Regulation: In astrocytes, GSTM1 modulates TNF-α-driven inflammatory pathways and fatty acid metabolism .

  • Therapeutic Target: Upregulation of GSTM1 via Nrf2 activation is a proposed strategy for CKD and neurodegenerative diseases .

Technical Considerations for Use

ParameterSpecification
Purity>95% (SDS-PAGE)
Activity AssayCDNB-glutathione conjugation at 25°C
StorageAvoid freeze-thaw cycles; use carrier proteins for long-term storage

Product Specs

Introduction
Glutathione S-transferases (GSTs) are a family of enzymes that play a crucial role in detoxification by catalyzing the conjugation of glutathione to a wide variety of electrophilic substrates, including carcinogens, therapeutic drugs, and products of oxidative stress. Among the various classes of GSTs, the mu class, encoded by genes located on chromosome 1p13.3, is highly polymorphic. These genetic variations can influence an individual's susceptibility to carcinogens and their response to certain drugs. Notably, individuals with null mutations in the mu class GST genes may exhibit an increased risk of developing certain types of cancer. The association between GST polymorphisms and cancer risk highlights the significance of these enzymes in cellular defense mechanisms and their potential as biomarkers for disease susceptibility.
Description
Recombinant GSTM1 from mouse, expressed in E. coli, is a purified protein. It is a single, non-glycosylated polypeptide chain with 238 amino acids (amino acids 1-218) and a molecular weight of 28.1 kDa. A 20 amino acid His-tag is present at the N-terminus of the protein. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The GSTM1 protein is supplied in a solution containing 20 mM Tris-HCl buffer at pH 8.0, 1 mM DTT (dithiothreitol), and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure long-term stability during frozen storage, it is advisable to add a carrier protein such as HSA (human serum albumin) or BSA (bovine serum albumin) to a final concentration of 0.1%. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the GSTM1 protein is determined to be greater than 95.0% using SDS-PAGE analysis.
Biological Activity
The specific activity of the GSTM1 enzyme is measured to be less than 11 units per milligram of protein. This activity is defined as the amount of enzyme required to catalyze the conjugation of 1.0 micromole of 1-chloro-2,4-dinitrobenzene (CDNB) with reduced glutathione per minute at a pH of 6.5 and a temperature of 25°C.
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

MGSSHHHHHH SSGLVPRGSH 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 a phase II detoxification enzyme involved in xenobiotic metabolism. In mice, the GSTM1 gene is located on chromosome 3, spanning 5724 bp and comprising 8 exons that encode a protein of 244 amino acids . The protein is expressed in various tissues including brain, liver, and kidneys.

The primary functions of GSTM1 include:

  • Catalyzing the conjugation of glutathione to electrophilic compounds for detoxification

  • Providing cellular protection against oxidative stress

  • Metabolizing environmental toxins and drugs

  • Regulating inflammatory responses, particularly in brain tissue where it promotes pro-inflammatory mediator production by astrocytes and enhances microglial activation

Recent research has also identified tissue-specific roles, including influence on neuroinflammatory processes in the brain where GSTM1 expression increases in the frontal cortex and hippocampus of aging mice , and potential involvement in kidney function, particularly in response to nephrotoxic agents like cisplatin .

How is GSTM1 gene knockout achieved in mouse models?

The most effective approach for generating GSTM1 knockout (GSTM1-KO) mice uses CRISPR-Cas9 gene editing technology. The process involves:

  • Target selection: Exon 2 of GSTM1 (sequence: GATCCGCATGCTCCTGGAAT) has proven effective as a specific target for knockout .

  • Guide RNA design: Design specific sgRNAs targeting the selected sequence in exon 2.

  • Microinjection: Inject Cas9 mRNA and sgRNAs into fertilized mouse oocytes.

  • Embryo transfer: Transfer injected embryos to pseudopregnant females.

  • Founder screening: Screen offspring DNA for mutations in the target region.

  • Breeding strategy: Breed heterozygous founders carrying desired mutations to establish homozygous knockout lines.

Importantly, since the knockout gene fragments are typically small (often just 5-13 bp), conventional agarose gel electrophoresis cannot differentiate between homozygous, heterozygous, and wild-type genotypes. Therefore, PCR amplification products should be sequenced and compared with wild-type sequences to confirm successful deletion of target bases .

What verification methods should be used to confirm successful GSTM1 knockout in mice?

A comprehensive verification approach requires multiple techniques to confirm successful GSTM1 knockout:

  • DNA sequencing: PCR amplification of the target region followed by sequencing to confirm deletion of target bases in GSTM1. This is essential since conventional gel electrophoresis cannot detect small deletions .

  • mRNA expression analysis: Real-time PCR to quantify GSTM1 mRNA levels in tissues of interest, comparing knockout mice to wild-type controls .

  • Protein expression analysis:

    • Western blotting to confirm reduced protein expression in target tissues

    • Immunohistochemistry and immunofluorescence staining to visualize absence of GSTM1 protein in tissue sections

  • Functional assays: Measuring GST enzyme activity in tissue homogenates to confirm functional consequences of the knockout.

  • Phenotypic analysis: Assessing whether the knockout results in expected phenotypic changes based on known GSTM1 functions, though single knockouts may not show obvious structural changes in tissues like kidney under normal conditions .

How does GSTM1 knockdown differ from null genotypes in experimental contexts?

Understanding the distinction between GSTM1 null genotypes and knockdown is crucial for experimental design:

GSTM1 null genotype:

  • Represents complete absence of the functional gene, either naturally occurring or experimentally induced

  • Results in complete absence of GSTM1 protein production

  • Is a permanent, heritable genetic condition

  • In mice, typically achieved through CRISPR-Cas9-mediated knockout of critical exons

GSTM1 knockdown:

  • Reflects temporary, partial reduction in gene expression

  • Typically achieved using RNA interference techniques, such as short hairpin RNA (shRNA) delivered via lentiviral vectors

  • Results in significantly reduced but not completely eliminated GSTM1 expression

  • Is reversible and can be regulated in intensity

  • Can be tissue-specific when appropriate promoters are used, such as astrocyte-specific knockdown achieved using AAV vectors with GSTM1 shRNAmir

Each approach has distinct advantages: null genotypes are valuable for studying complete loss-of-function, while knockdown approaches permit tissue-specific, time-controlled, and dose-dependent reduction in gene expression. For example, GSTM1 knockdown in astrocytes has been used to investigate its role in neuroinflammation while maintaining normal expression in other cell types .

What are the optimal storage and handling conditions for recombinant His-tagged GSTM1 protein?

For optimal activity and stability of recombinant His-tagged GSTM1 protein:

Storage conditions:

  • Lyophilized protein: Store at -20°C to -80°C, where it remains stable for up to 12 months

  • Reconstituted protein solution: Can be stored at 4-8°C for 2-7 days

  • Longer-term storage of reconstituted protein: Prepare aliquots and store at < -20°C, where they remain stable for approximately 3 months

  • Avoid repeated freeze-thaw cycles which lead to protein denaturation and activity loss

Reconstitution protocol:

  • Reconstitute lyophilized protein in sterile PBS (pH 7.4)

  • Most recombinant GSTM1 preparations contain 5-8% trehalose as a stabilizing agent

  • Allow protein to sit for 10-20 minutes at room temperature after adding buffer to ensure complete solubilization

  • Gently pipette or swirl to mix; avoid vigorous vortexing which can denature the protein

Quality control considerations:

  • Verify protein purity by SDS-PAGE (should be >95%)

  • Test enzymatic activity using standard GST substrates such as 1-chloro-2,4-dinitrobenzene (CDNB)

  • For immunological applications, confirm immunoreactivity before proceeding with critical experiments

  • Consider that the His-tag (typically N-terminal for mouse GSTM1 ) may influence binding characteristics in some experimental contexts

What phenotypic differences exist between GSTM1, GSTT1, and GSTM1/GSTT1 double knockout mouse models?

Comparative analysis of GST knockout models reveals distinct and overlapping phenotypes:

GSTM1 knockout (GSTM1-KO) mice:

  • Under normal physiological conditions, show no obvious structural abnormalities in kidney tissue

  • Demonstrate normal kidney function parameters including blood urea nitrogen (BUN) and creatinine (CREA) levels

  • When challenged with cisplatin, show increased susceptibility to acute kidney injury compared to wild-type mice

  • May exhibit altered inflammatory responses, particularly in the central nervous system

GSTT1 knockout (GSTT1-KO) mice:

  • GSTT1 gene is located on chromosome 10, spanning 14,772 bp and consisting of 5 exons encoding 190 amino acids

  • Like GSTM1-KO mice, show normal kidney structure and function under baseline conditions

  • May exhibit differential responses to xenobiotics due to distinct substrate specificity

GSTM1/GSTT1 double knockout (Gstm1/Gstt1-DKO) mice:

  • Successfully generated by breeding GSTM1-KO and GSTT1-KO mice together

  • Maintain intact kidney structure and morphology despite absence of both enzymes

  • Show no significant differences in kidney-to-body weight ratio, BUN, or CREA compared to wild-type mice under normal conditions

  • Particularly valuable for research as they mirror a common genetic condition in human populations associated with increased disease risk

These findings suggest that while individual or combined loss of GSTM1 and GSTT1 may not cause overt abnormalities under normal conditions, their importance becomes apparent under stress conditions or xenobiotic challenges.

How does GSTM1 deficiency affect neuroinflammatory responses in mouse models?

GSTM1 deficiency significantly alters neuroinflammatory responses through several mechanisms:

Altered astrocyte activation profile:

  • GSTM1 knockdown in astrocytes downregulates pro-inflammatory gene expression while upregulating genes involved in interferon responses and fatty acid metabolism

  • Production of specific cytokines/chemokines (including CXCL1, CSF2, and CXCL2) is severely impaired in GSTM1-deficient astrocytes following TNF-α stimulation

  • CSF2, which is reduced in GSTM1-deficient astrocytes, normally contributes to microglial regulation and neuroprotection during inflammation

Effects on neuronal function:

  • Astrocyte-specific GSTM1 knockdown leads to reduced neuronal activity, as evidenced by decreased c-Fos expression in neurons following lipopolysaccharide (LPS) challenge

  • Paradoxically, GSTM1 reduction in astrocytes increases neuronal stress levels while attenuating neuronal activities during LPS-induced brain inflammation

  • This suggests GSTM1 in astrocytes maintains neuronal activities during inflammatory conditions through complex intercellular signaling

Transcriptional program alterations:

  • RNA-seq analysis reveals GSTM1 knockdown significantly affects TNF-α-dependent transcriptional programs in astrocytes

  • Gene Set Enrichment Analysis (GSEA) of differentially expressed genes shows altered pathway activation in response to pro-inflammatory stimuli

Age-related implications:

  • GSTM1 expression increases in the frontal cortex and hippocampus of aging mice

  • This age-related increase suggests GSTM1 may contribute to age-associated neuroinflammatory changes

These findings position GSTM1 as a potential therapeutic target for modulating neuroinflammation in various neurological conditions.

What methodological approaches are most effective for studying GSTM1's role in astrocyte-neuron interactions?

Several sophisticated methodological approaches have proven effective for investigating GSTM1's role in astrocyte-neuron interactions:

In vitro approaches:

  • Lentiviral-mediated knockdown in primary astrocyte cultures:

    • Primary mouse cortical astrocytes infected with lentivirus vectors encoding GSTM1 shRNA or non-silencing controls

    • Knockdown efficiency verified by qPCR and Western blotting

    • Stimulation with TNF-α (50 ng/ml for 6 hours) to induce inflammatory activation

    • RNA-seq analysis to assess transcriptome-wide changes in gene expression

    • Gene Set Enrichment Analysis (GSEA) to identify affected pathways

  • Co-culture systems:

    • Neuron-astrocyte co-cultures with GSTM1-deficient or control astrocytes

    • Assessment of neuronal viability, morphology, and activity markers

    • Transwell systems to distinguish between contact-dependent and soluble factor-mediated effects

In vivo approaches:

  • Astrocyte-specific GSTM1 knockdown:

    • Cre-dependent viral vector systems using GFAP-Cre mice for astrocyte-specific manipulation

    • Stereotactic injection of AAV vectors encoding GSTM1 or control shRNAmir into specific brain regions

    • Three-week post-injection period for optimal expression and knockdown

  • Inflammatory challenge protocols:

    • Systemic administration of LPS to induce neuroinflammation

    • Brain tissue collection at defined timepoints (e.g., 6 hours post-LPS)

    • Immunohistochemical assessment of neuronal activation using c-Fos as a marker

    • Analysis of microglial activation state in the vicinity of GSTM1-deficient astrocytes

These complementary approaches provide a comprehensive understanding of how GSTM1 in astrocytes influences neuronal function during inflammatory conditions, revealing both cell-autonomous effects in astrocytes and non-cell-autonomous effects on surrounding neurons.

How should researchers address contradictory findings regarding GSTM1's role in reproductive health?

Addressing contradictory findings regarding GSTM1's role in reproductive health requires systematic methodological approaches:

Reconciling unexpected results:

Some contradictory findings exist, such as the unexpected observation that the GSTT1 non-null genotype (normal enzyme levels) was associated with reduced sperm count and concentration, contrary to the hypothesis that GST enzymes would be protective for reproductive parameters . To address such contradictions:

  • Design comprehensive genotype-phenotype studies:

    • Include homozygous null, heterozygous, and wild-type genotypes

    • Assess multiple GST family members simultaneously (GSTM1, GSTT1, GSTZ1)

    • Use larger sample sizes to account for genetic heterogeneity (previous studies had 162 participants)

    • Apply appropriate statistical analysis with adjustment for confounders (race/ethnicity, age, study site)

  • Incorporate exposure assessment:

    • Previous research was limited by insufficient sample size to examine interactions with environmental exposures

    • Design studies with adequate power to detect gene-environment interactions

    • Include detailed exposure assessment for toxicants metabolized by GST enzymes

    • Consider relevant exposure windows (e.g., 90 days for spermatogenesis)

  • Standardize outcome measurements:

    • Implement consistent data transformations (e.g., natural log transformation for sperm count and concentration)

    • Standardize sample collection protocols (consistent abstinence periods)

    • Include multiple outcome measures beyond conventional semen parameters

    • Consider functional sperm assessments like DNA fragmentation index

  • Apply advanced molecular approaches:

    • Conduct targeted pathway analysis focusing on reproductive signaling networks

    • Consider compensatory mechanisms that may mask effects in single-gene studies

    • Utilize newer genomic technologies to complement traditional genotyping approaches

By implementing these methodological improvements, researchers can better understand the true relationship between GSTM1 polymorphisms and reproductive health, resolving apparent contradictions in the current literature.

What experimental design considerations are crucial when studying interactions between GSTM1 polymorphisms and environmental toxins?

When investigating interactions between GSTM1 polymorphisms and environmental toxins, several critical experimental design considerations should be addressed:

Animal model selection:

  • Appropriate genetic models:

    • Use CRISPR-Cas9 generated GSTM1 knockout mice that precisely recapitulate human polymorphisms

    • Consider double knockout models (e.g., GSTM1/GSTT1-DKO) to mirror common human genetic variations

    • Include heterozygous models to study gene-dose effects

  • Exposure paradigms:

    • Design both acute and chronic exposure protocols

    • Include recovery periods to assess reversibility

    • Use environmentally relevant dosing regimens

    • Consider developmental exposures during critical windows

  • Multi-tissue assessment:

    • Evaluate effects in multiple organs where GSTM1 is expressed

    • Consider barrier tissues where detoxification is critical

    • Examine tissue-specific differences in response to the same toxicant

Human studies design:

  • Sampling strategy:

    • Ensure adequate representation of all genotypes

    • Calculate appropriate sample sizes for gene-environment interactions

    • Use appropriate biological sampling methods (e.g., buccal cells for DNA extraction)

    • Consider potential selection bias in participant recruitment

  • Confounder control:

    • Identify and measure potential confounders specific to the toxicant and outcome

    • Account for demographic variables (age, race/ethnicity)

    • Consider study site as a potential confounder in multi-center studies

    • Adjust for other genetic variations in metabolic pathways

  • Statistical approaches:

    • Use appropriate models for interaction testing

    • Address multiple comparison issues when assessing numerous toxicants or outcomes

    • Consider non-linear exposure-response relationships

    • Implement stratified analyses to examine effects within genotype subgroups

By addressing these experimental design considerations, researchers can generate more robust findings regarding interactions between GSTM1 polymorphisms and environmental toxins, leading to better understanding of susceptibility factors for toxicant-induced diseases.

What are the optimal approaches for studying GSTM1 protein-protein interactions using His-tagged recombinant proteins?

Studying GSTM1 protein-protein interactions using His-tagged recombinant proteins requires careful methodological consideration:

Protein preparation and characterization:

  • Expression and purification:

    • E. coli expression systems are commonly used for mouse GSTM1 production

    • Utilize nickel or cobalt affinity chromatography for His-tag capture

    • Implement secondary purification steps to ensure >95% purity

    • Verify structural integrity using biophysical techniques

  • Tag considerations:

    • N-terminal His-tags are common for GSTM1 expression

    • Evaluate whether the tag affects protein function through activity assays

    • Consider tag removal using specific proteases for interaction studies where the tag might interfere

    • Mouse GSTM1 with His-tag has a molecular mass of approximately 28 kDa

Interaction analysis techniques:

  • Affinity-based methods:

    • Immobilize His-tagged GSTM1 on Ni-NTA resin for pull-down assays

    • Include appropriate controls (non-specific His-tagged proteins, tag-only controls)

    • Use Surface Plasmon Resonance (SPR) with immobilized His-tagged GSTM1 for kinetic analysis

    • Apply AlphaScreen/AlphaLISA for detecting interactions without separation steps

  • Solution-based methods:

    • Isothermal Titration Calorimetry (ITC) for direct measurement of binding thermodynamics

    • Microscale Thermophoresis (MST) for detecting interactions with small sample volumes

    • Size exclusion chromatography combined with multi-angle light scattering to determine complex formation

  • Validation approaches:

    • Measure GSTM1 enzymatic activity using standard substrates in the presence of interaction partners

    • Develop cellular validation using co-immunoprecipitation or proximity ligation assays

    • Apply mutagenesis to identify critical residues in the interaction interface

These complementary approaches provide a comprehensive strategy for characterizing GSTM1 protein-protein interactions, from initial screening to detailed mechanistic understanding, using His-tagged recombinant proteins as valuable research tools.

Product Science Overview

Glutathione S-Transferase M1 (GSTM1)

GSTM1 is one of the isoforms of GST and is encoded by the GSTM1 gene. This enzyme is particularly important in the metabolism of carcinogens, drugs, and products of oxidative stress. The GSTM1 gene is polymorphic, meaning that there are variations in the gene sequence among individuals, which can affect the enzyme’s activity and, consequently, an individual’s susceptibility to certain diseases.

Recombinant GSTM1

Recombinant GSTM1 refers to the enzyme produced through recombinant DNA technology. This involves inserting the GSTM1 gene into an expression vector, which is then introduced into a host organism, such as E. coli, to produce the enzyme in large quantities. The recombinant enzyme can be tagged with a histidine tag (His Tag) to facilitate its purification and detection.

His Tag

A His Tag is a string of histidine residues (usually six) added to either the N- or C-terminus of a protein. This tag allows for easy purification of the recombinant protein using metal affinity chromatography, as histidine residues have a high affinity for divalent metal ions like nickel or cobalt. The His Tag also aids in the detection of the protein using anti-His antibodies.

Production and Purification

The production of recombinant GSTM1 involves cloning the GSTM1 gene into an expression vector that includes a His Tag sequence. The vector is then introduced into a host organism, such as E. coli, where the recombinant protein is expressed. The His Tag allows for the purification of the protein using nickel or cobalt affinity chromatography .

Applications

Recombinant GSTM1 with a His Tag is widely used in research to study the enzyme’s function, structure, and interactions with other molecules. It is also used in drug development to screen for potential inhibitors of GSTM1, which could be used to enhance the efficacy of chemotherapy drugs by preventing the detoxification of these drugs by GSTM1.

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.