GCLM antibodies are validated for multiple techniques, with optimized dilutions for each:
KO Testing: Antibodies like ab126704 (Abcam) show no signal in GCLM KO HAP1 cells, confirming specificity .
Cross-reactivity: Tested in human liver, lung cancer tissues, and rodent cell lines .
GCLM antibodies have elucidated the protein’s role in tumor progression and immune evasion:
GCLM Overexpression in Bladder Cancer (BCa):
Ferroptosis Regulation:
Immune Cell Correlations:
Immune Checkpoint Links:
GCLM is identified as a latent therapeutic target:
Targeting GCLM: May enhance immune checkpoint blockade (ICB) efficacy by modulating tumor immune microenvironment (TIME) .
Prognostic Biomarker: High GCLM expression predicts poor prognosis in BCa .
CHO Cell Engineering: Overexpressing GCLM (but not GCLC) in CHO cells increases monoclonal antibody titers by 70%, despite unchanged GSH levels .
Mechanism: GCLM may regulate cellular processes beyond GSH synthesis, such as protein folding or redox balance .
A selection of commercially available antibodies is summarized below:
GCLM is the regulatory subunit of glutamate-cysteine ligase, also known as gamma-glutamylcysteine synthetase, which serves as the first rate-limiting enzyme in glutathione synthesis. Glutathione plays a crucial role as a vital antioxidant that protects cells from oxidative stress and maintains cellular redox balance. GCLM forms a heterodimer with the heavy catalytic subunit, and their interaction is regulated through the formation of a reversible disulfide bond that enhances enzyme activity . The human GCLM gene is located on chromosome 1p22-p21, while the catalytic subunit gene is found on chromosome 6p12 . Research on GCLM is significant for understanding cellular defense mechanisms against oxidative damage and has implications in numerous pathological conditions.
Multiple types of GCLM antibodies have been developed to suit various research needs:
Additionally, conjugated versions are available for specialized applications, including HRP-conjugated, FITC-conjugated, and PE-conjugated antibodies for detection without secondary antibodies .
Based on the search results, GCLM antibodies have been validated for numerous applications:
Western blotting (WB): All examined antibodies are validated for WB with dilution ranges typically between 1:500-1:2000
Immunocytochemistry/Immunofluorescence (ICC/IF): Multiple antibodies show successful application
Immunohistochemistry (IHC-P): Several antibodies are validated for paraffin-embedded tissues
Immunoprecipitation (IP): Select antibodies are confirmed effective
Additional specialized applications: Some antibodies have been validated for ChIP, ChIP-seq, RIP, Flow Cytometry, and ELISA
Selection should be based on several critical factors:
Species reactivity: Ensure compatibility with your experimental model (human, mouse, rat)
Application requirements: Different antibodies perform optimally in specific applications; for instance, monoclonal antibodies like EPR6667 demonstrate superior specificity for IP and IHC-P applications
Clonality considerations: Polyclonal antibodies often provide higher sensitivity but potentially lower specificity compared to monoclonals
Validation status: Prioritize antibodies with knockout (KO) testing or extensive citation records
Epitope location: Consider whether your research question requires detection of specific regions of GCLM
Signal strength requirements: For detecting low-abundance GCLM expression, higher-sensitivity detection systems or brighter fluorophore conjugates may be necessary
When studying post-translational modifications or protein-protein interactions, special consideration should be given to epitope accessibility in the experimental conditions.
Rigorous controls are critical for meaningful GCLM antibody experiments:
Positive controls: Cell lines or tissues with confirmed GCLM expression (e.g., A431, C8D30, rat liver)
Negative controls: Consider GCLM knockout cells or tissues, or primary antibody omission controls
Loading controls: Essential for quantitative western blot analysis
Isotype controls: Particularly important for flow cytometry and immunohistochemistry applications to distinguish non-specific binding
Blocking peptide competition: Can confirm antibody specificity by pre-incubating with the immunizing peptide
Transfection controls: Non-transfected versus transfected samples can validate antibody specificity, as demonstrated with the GTX114075 antibody on HeLa extracts
For flow cytometry applications, fluorescence minus one (FMO) controls are recommended to establish proper gating strategies and account for spillover when using multiple fluorophores .
Optimal sample preparation varies by application:
For Western Blotting:
Standard SDS-PAGE (12% gel) has been validated for GCLM detection
Typical protein loading: 30-50 μg of whole cell lysate or tissue extract
Expected molecular weight: 31 kDa (calculated MW: 28-30 kDa)
Buffer considerations: PBS with 0.02% sodium azide, 50% glycerol, pH 7.3 has been used successfully for antibody storage
For Immunohistochemistry:
Heat-mediated antigen retrieval using Tris-EDTA buffer (pH 9.0) is recommended
For monoclonal antibodies like EPR6667, dilution of approximately 1:50 (2.4 μg/mL) has been validated
For Immunofluorescence:
Methanol fixation has been documented for successful GCLM antibody staining
Cellular localization should be predominantly cytoplasmic
Several strategies can address inconsistent detection:
Antibody titration: Determine optimal concentration through titration experiments rather than relying solely on manufacturer recommendations
Sample preparation optimization: Ensure complete protein denaturation and consider phosphatase/protease inhibitors to prevent degradation
Transfer efficiency verification: Use reversible staining methods to confirm successful protein transfer
Signal enhancement techniques: Consider enhanced chemiluminescence systems for low abundance detection
Membrane selection: PVDF membranes may provide better protein retention than nitrocellulose for some applications
Blocking optimization: Test different blocking agents (milk vs. BSA) as milk may contain phosphatases that could interfere with detection of phosphorylated GCLM forms
If primary issues persist, comparing results across multiple GCLM antibodies targeting different epitopes can help validate findings and resolve inconsistencies.
High background can compromise data interpretation in immunostaining:
Antibody dilution optimization: Increasing dilution factors beyond manufacturer recommendations may reduce non-specific binding
Enhanced blocking protocols: Extending blocking time or using alternative blocking agents can reduce background
Fc receptor blocking: Particularly important in immune cell studies to prevent non-specific binding
Secondary antibody cross-reactivity: Test secondary antibodies alone to identify potential direct binding to samples
Autofluorescence reduction: Additional washing steps, shorter incubation times, or specialized quenching reagents
Dead cell exclusion: Implement viability dyes as dead cells bind antibodies non-specifically, particularly in flow cytometry applications
For flow cytometry specifically, creating dump channels can improve resolution by excluding unwanted cell populations .
Variations in banding patterns may result from:
Post-translational modifications: Phosphorylation, ubiquitination, or other modifications can alter migration patterns
Alternative splicing: Multiple transcript variants of GCLM have been reported
Protein-protein interactions: Incomplete denaturation may result in higher molecular weight complexes
Proteolytic processing: Sample handling can lead to degradation products
Experimental conditions: Variations in gel percentage, running conditions, or buffer systems
When unexpected bands appear, verification through additional techniques (IP-Western, mass spectrometry, or RNA interference) can help determine their identity and relevance to GCLM biology.
GCLM antibodies enable several advanced research approaches in oxidative stress studies:
Stress response profiling: Tracking GCLM protein expression changes under various oxidative challenges
Subcellular localization dynamics: Using immunofluorescence to monitor potential translocation events during stress responses
Correlation studies: Combining GCLM detection with glutathione measurement assays to establish functional relationships
Interaction mapping: Employing co-immunoprecipitation with GCLM antibodies to identify novel protein interactions in stress conditions
Tissue-specific expression analysis: IHC application in disease models to identify altered regulation across tissues
These approaches can be particularly valuable in models of diseases associated with redox imbalance, including neurodegenerative conditions, cancer, and cardiovascular disorders.
Several sophisticated experimental approaches can be implemented:
Promoter-binding studies: Combining ChIP assays with GCLM antibodies to investigate transcriptional regulation
Post-translational modification mapping: Using modification-specific antibodies alongside total GCLM antibodies
Structure-function analysis: Correlating structural changes in GCLM-GCLC interaction with enzyme activity measurements
Compartmentalization studies: Fractionation experiments with GCLM antibodies to track subcellular distribution under various conditions
Dynamic interaction studies: FRET or BRET approaches using labeled antibodies or fusion proteins to monitor real-time interactions
These approaches can reveal regulatory mechanisms that may serve as intervention points for diseases associated with glutathione depletion.
Designing effective multi-parameter panels requires careful consideration:
Antigen density matching: Pair GCLM antibody conjugates with appropriate fluorophores based on expected expression levels; brighter fluorophores should be reserved for lower-expressed markers
Spectral compatibility: Separate fluorophores across different lasers and filters to minimize compensation requirements
Panel complexity management: When designing complex panels including GCLM, use the pre-loaded cytometer settings in panel builders to ensure compatibility
Co-expression analysis: Design panels to simultaneously evaluate GCLM with related proteins in the glutathione synthesis pathway
Cell subset identification: Include lineage markers alongside GCLM to identify cell-type specific expression patterns
For rare cell populations expressing GCLM, collection of significantly more cells may be necessary to obtain statistically meaningful data .
Proper interpretation of tissue-specific GCLM expression requires:
Normalization strategies: Careful selection of reference genes/proteins that maintain stable expression across compared tissues
Quantification methods: Densitometry for Western blots or digital pathology tools for IHC should be standardized
Statistical approaches: Appropriate statistical tests based on data distribution and experimental design
Validation across techniques: Confirmation of expression differences using complementary methods (qPCR, Western blot, IHC)
Functional correlation: Relating expression differences to glutathione levels or oxidative stress markers
Immunohistochemistry results demonstrate varied GCLM expression across tissues, with notable expression in liver and cardiac muscle, reflecting tissue-specific requirements for glutathione synthesis capacity .
When facing contradictory results:
Epitope mapping: Determine if antibodies recognize different regions that may be differentially accessible
Validation through knockdown/knockout: siRNA or CRISPR approaches can confirm specificity
Cross-platform verification: Apply multiple detection methods (e.g., mass spectrometry) for confirmation
Isoform-specific analysis: Consider whether antibodies might differentially detect splice variants
Technical replication: Systematic troubleshooting with consistent protocols across laboratories
Literature comparison: Review published findings for consensus patterns
Careful documentation of experimental conditions, antibody lot numbers, and exact protocols can help identify sources of variability.
Post-translational modifications present important considerations:
Epitope masking: Phosphorylation, acetylation, or other modifications may alter antibody binding efficiency
Conformation changes: Modifications can induce structural changes affecting epitope accessibility
Functional correlation: Specific modifications may correlate with altered GCLM-GCLC interaction or enzyme activity
Dynamic regulation: Temporal changes in modification status may explain variable detection
Specialized antibodies: Consider using modification-specific antibodies alongside total GCLM antibodies
The documented reversible disulfide bond formation between GCLM and the catalytic subunit represents a key post-translational regulatory mechanism that enhances enzyme activity , highlighting the importance of considering redox conditions in experimental design and interpretation.
Glutamate-Cysteine Ligase (GCL) is a critical enzyme in the biosynthesis of glutathione, a major antioxidant in cellular defense mechanisms. The enzyme is composed of two subunits: the catalytic subunit (GCLC) and the modifier subunit (GCLM). The modifier subunit, GCLM, enhances the catalytic efficiency of GCLC, forming a functional holoenzyme complex. This article delves into the background of the GCLM, particularly focusing on the mouse anti-human variant.
GCLM is a protein-coding gene that plays a pivotal role in glutathione synthesis. The enzyme’s activity is crucial for maintaining cellular redox balance and protecting cells from oxidative stress. GCLM, in conjunction with GCLC, catalyzes the first and rate-limiting step in glutathione biosynthesis, which involves the ligation of glutamate and cysteine to form γ-glutamylcysteine .
The GCLM subunit is essential for the optimal functioning of the GCL enzyme. It enhances the catalytic activity of GCLC, thereby increasing the overall efficiency of glutathione production. Glutathione is vital for detoxifying reactive oxygen species (ROS) and maintaining cellular homeostasis. Deficiencies in GCLM can lead to reduced glutathione levels, resulting in increased susceptibility to oxidative stress and related pathologies .
Mouse anti-human GCLM antibodies are monoclonal antibodies developed to specifically target the human GCLM protein. These antibodies are used in various research applications, including Western blotting, immunoprecipitation, and immunohistochemistry. They are valuable tools for studying the expression and function of GCLM in different biological contexts .
Research involving GCLM has significant implications for understanding oxidative stress-related diseases. Studies using GCLM knockout mice have shown that the absence of this subunit leads to decreased glutathione levels and increased oxidative damage in tissues. This model has been instrumental in elucidating the role of GCLM in various physiological and pathological processes .