GCLC encodes the catalytic subunit of glutamate-cysteine ligase (GCL), the enzyme responsible for the ATP-dependent condensation of glutamate and cysteine to form γ-glutamylcysteine—the first step in GSH synthesis . GCLC functions as a heterodimer with its regulatory subunit GCLM, enhancing catalytic efficiency and reducing feedback inhibition by GSH . Dysregulation of GCLC is linked to pathologies such as cancer, liver fibrosis, and neurodegenerative diseases .
GCLC antibodies are protein-specific reagents designed to detect and quantify GCLC expression in research settings. They are critical for:
Localization studies: Identifying cytoplasmic and nuclear GCLC distribution .
Expression profiling: Assessing GCLC levels in diseases like hepatocellular carcinoma (HCC) and hepatitis C virus (HCV)-related fibrosis .
Mechanistic studies: Investigating post-translational modifications and holoenzyme dynamics .
Cancer Prognosis: High GCLC expression in HCC tumors correlates with poor survival (median OS: 32 vs. 74 months, P < 0.05) .
Liver Fibrosis: GCLC overexpression in hepatic stellate cells (HSCs) reduces ROS and suppresses activation markers (α-SMA, COL1) .
Oxidative Stress: Post-translational activation of GCL holoenzyme occurs within minutes of oxidative insult, independent of disulfide bonds .
Validation: Antibodies like ab207777 show specificity via siRNA knockdown and immunoprecipitation .
Buffer Compatibility: Optimal performance in 5% non-fat dry milk/TBST .
Cross-Reactivity: Some antibodies (e.g., 12601-1-AP) detect goat GCLC, expanding translational research utility .
Applications : Western blot assays
Sample type: Human cells
Review: In parallel, the effects of SHQA were also observed for Nrf2-driven expression of antioxidant/phase II detoxifying enzymes, including HO-1, NQO1, GCLc, GCLm, and TrxR. In particular, HO-1 expression significantly increased after pretreatment with SHQA at 0.1 µM.
GCLC (Glutamate-Cysteine Ligase, Catalytic Subunit) is the first rate-limiting enzyme in glutathione (GSH) biosynthesis, catalyzing the ATP-dependent ligation of L-glutamate and L-cysteine . GCLC antibodies are crucial research tools because:
They enable detection and quantification of GCLC protein expression in various tissues and cell types
They help investigate glutathione synthesis regulation, which is critical in oxidative stress responses
They allow researchers to study mechanisms behind conditions like hemolytic anemia (HAGGSD) caused by GCLC defects
They facilitate research on the association between GCLC gene expression and coronary endothelial vasomotor dysfunction and myocardial infarction
GCLC antibodies provide a methodological approach to visualizing and measuring this protein across multiple experimental platforms including Western blotting, immunohistochemistry, and immunofluorescence.
GCLC antibodies are versatile tools with multiple validated applications:
For optimal results, researchers should validate each antibody for their specific application and experimental conditions, as performance can vary between different antibody clones and preparations .
GCLC antibodies display varying species reactivity profiles, which is crucial to consider when selecting the appropriate antibody:
Human GCLC shares approximately 94% amino acid sequence identity with both mouse and rat GCLC , explaining the broad cross-reactivity observed with many antibodies. Researchers should verify the reactivity for their specific model organism, particularly when working with less common species.
Proper storage and handling of GCLC antibodies is essential for maintaining their performance and extending their useful life:
Storage temperature: Most GCLC antibodies should be stored at -20°C for long-term stability
Aliquoting recommendations: Aliquot antibodies to avoid repeated freeze-thaw cycles, which can degrade antibody quality
Buffer composition: Typically stored in PBS with additives such as:
Thawing protocol: Thaw antibodies on ice or at 4°C, and centrifuge briefly before use
Shelf life considerations: Many GCLC antibodies have a guaranteed shelf life of 12 months from date of dispatch when properly stored
For unconjugated GCLC antibodies, avoid repeated freeze-thaw cycles as this significantly reduces antibody efficacy. Some manufacturers specifically note that aliquoting is unnecessary for -20°C storage in their specific formulation , so always follow vendor-specific recommendations.
The choice between monoclonal and polyclonal GCLC antibodies significantly impacts experimental outcomes:
For detecting post-translational modifications or when working in denaturing conditions, polyclonal antibodies often provide advantages. Conversely, when precise epitope targeting or reproducibility across experiments is crucial, monoclonal antibodies are preferable, particularly recombinant monoclonals that offer enhanced consistency .
Achieving optimal results with GCLC antibodies in Western blotting requires careful attention to protocol details:
Sample preparation:
Electrophoresis and transfer parameters:
Blocking and antibody incubation:
Detection and troubleshooting:
Both chemiluminescence and fluorescence detection systems work well
If background is high, increasing blocking time or adjusting antibody concentration can help
If signal is weak, longer exposure times or increased antibody concentration may be necessary
The wide range of recommended dilutions (1:1000-1:50000) highlights the importance of empirical optimization for each specific antibody and experimental system .
Rigorous validation of GCLC antibodies ensures experimental reliability and reproducibility:
Knockout/knockdown validation:
Multiple antibody approach:
Immunogen peptide blocking:
Cross-platform validation:
Observed molecular weight verification:
In publications, researchers should report how antibody specificity was validated to ensure experimental reproducibility.
The epitope specificity of GCLC antibodies significantly impacts their performance across different applications:
The choice of epitope region impacts:
Structural recognition: Antibodies targeting regions involved in protein-protein interactions may show reduced binding when GCLC is in complexes
Post-translational modification detection: Modifications near the epitope may block antibody binding
Isoform detection: Different epitopes may be present or absent in specific isoforms
Functional domain recognition: Antibodies targeting functional domains can provide insights into activity-structure relationships
For comprehensive studies, using antibodies targeting different epitopes provides more complete information about GCLC biology and potential modifications .
The integration of GCLC antibody research with advanced computational methods represents an emerging frontier:
Deep learning in antibody design:
Machine learning for epitope prediction:
Computational prediction of optimal GCLC epitopes could improve antibody design
In-silico approaches may identify epitopes that are:
Highly specific to GCLC
Accessible in native protein conformations
Conserved across species for broad reactivity
Automated image analysis in IHC/IF:
Deep learning algorithms can quantify GCLC expression patterns in tissue sections
Reduces subjectivity in image interpretation and increases throughput
Integrated multi-omics approaches:
Combining antibody-based protein detection with transcriptomics and metabolomics
Machine learning algorithms can identify patterns across datasets that might be missed by conventional analysis
The field is progressing toward "in-silico discovery of antibody-based biotherapeutics" that could "accelerate in-silico discovery... and expand the druggable antigen space" , which may eventually include novel GCLC-targeted approaches.
When facing inconsistent results with GCLC antibodies, systematic troubleshooting approaches are essential:
Sample preparation variables:
Cell/tissue lysis conditions affect protein extraction efficiency
Protein degradation during preparation can be prevented with:
Fresh protease inhibitors
Maintaining samples at 4°C
Avoiding repeated freeze-thaw cycles of lysates
Protocol optimization strategies:
Antibody selection considerations:
Technical validation approaches:
For reproducible results, researchers should meticulously document all experimental conditions and antibody details (including catalog number, lot, and dilution) in their methods sections.
The selection of appropriate GCLC antibody format should be guided by experimental requirements:
When selecting a GCLC antibody, researchers should consider:
Required application sensitivity (polyclonals typically offer higher sensitivity)
Need for reproducibility across experiments (monoclonals or recombinants provide greater consistency)
Target species (check validated reactivity profiles)
Epitope accessibility in your experimental system
Validation data availability for your specific application
For cutting-edge applications like super-resolution microscopy or in vivo imaging, specialized antibody formats may be required that offer optimal signal-to-noise ratios in these demanding contexts.
Successful immunohistochemistry with GCLC antibodies requires attention to several critical parameters:
Tissue preparation and fixation:
Antigen retrieval optimization:
Blocking and antibody incubation:
Detection system selection:
Amplification systems (e.g., TSA) may be needed for low expression targets
Chromogenic vs. fluorescent detection depends on:
Need for co-localization studies
Tissue autofluorescence concerns
Quantification requirements
Validation and controls:
Include tissue with known GCLC expression patterns as positive control
Omission of primary antibody serves as a technical negative control
Ideally, include GCLC-knockout or knockdown samples as biological negative controls
The statement "It is recommended that this reagent should be titrated in each testing system to obtain optimal results" emphasizes the importance of empirical optimization for specific experimental conditions .
Multiplex detection involving GCLC antibodies requires careful planning and optimization:
Antibody compatibility considerations:
Select GCLC antibodies from different host species than other target antibodies
Alternatively, use directly conjugated primary antibodies to avoid cross-reactivity
Consider isotype-specific secondary antibodies when using multiple primary antibodies from the same species
Sequential staining protocols:
If using antibodies from the same species, employ sequential staining with:
Complete elution between antibodies, or
Blocking of the first primary antibody before applying the second
Fluorophore selection strategies:
Choose fluorophores with minimal spectral overlap
Account for tissue autofluorescence when selecting fluorophores
Consider signal intensity when pairing fluorophores with targets (brighter fluorophores for less abundant targets)
Spatial resolution optimization:
For subcellular co-localization studies, confocal or super-resolution microscopy may be required
Antibody penetration into tissue sections may require optimization of incubation times and detergent concentrations
Data analysis approaches:
Use spectral unmixing algorithms to separate overlapping fluorophore signals
Quantify co-localization using appropriate statistical methods
Consider automated image analysis for unbiased quantification
When designing multiplex experiments, researchers should first validate each antibody individually before combining them to ensure specific detection of each target.
Investigating GCLC in oxidative stress contexts requires careful experimental design:
Oxidative stress induction methods:
Chemical inducers (H₂O₂, paraquat, menadione)
Metabolic inducers (high glucose, hypoxia/reoxygenation)
Environmental stressors (UV, radiation, heavy metals)
Each method produces distinct patterns of oxidative damage and stress response
Temporal considerations:
GCLC expression changes dynamically after oxidative stress
Include multiple time points (early, intermediate, late responses)
Consider both acute and chronic oxidative stress models
Controls and validation:
Include antioxidant treatment controls
Measure multiple oxidative stress markers (ROS levels, lipid peroxidation, protein carbonylation)
Confirm GCLC functional changes (glutathione synthesis rates, GSH/GSSG ratio)
Complementary approaches:
Combine protein detection (using GCLC antibodies) with:
mRNA expression analysis
Enzyme activity assays
Glutathione level measurements
Assessment of downstream antioxidant responses
Cellular compartmentalization analysis:
Examine potential GCLC translocation between subcellular compartments
Use appropriate cellular fractionation techniques
Employ IF with co-localization markers for different organelles
When interpreting results, consider that GCLC regulation occurs at multiple levels (transcriptional, post-transcriptional, post-translational) and may differ between acute and chronic stress conditions.
Recent breakthroughs in computational antibody design present exciting opportunities for next-generation GCLC antibodies:
Deep learning approaches:
In silico epitope mapping:
Computational prediction of immunodominant GCLC epitopes
Identification of conserved epitopes across species for broader cross-reactivity
Prediction of epitopes that remain accessible in different protein conformations
Structure-guided antibody engineering:
Using AlphaFold2 and similar AI systems to predict GCLC structure
Design of antibodies targeting specific functional domains
Engineering antibodies with enhanced affinity and specificity
High-throughput screening integration:
Combining computational design with high-throughput experimental validation
Machine learning algorithms to predict antibody performance from sequence data
Iterative design-build-test cycles for rapid antibody optimization
The recent development of "a deep learning model for computationally generating libraries of highly human antibody variable regions" demonstrates the potential of these approaches to revolutionize antibody development, potentially including next-generation GCLC antibodies with superior performance profiles.
GCLC antibodies are becoming increasingly valuable tools in elucidating disease mechanisms:
Neurodegenerative diseases:
Tracking GCLC expression changes in Alzheimer's, Parkinson's, and ALS models
Investigating the role of glutathione depletion in neuronal vulnerability
Monitoring GCLC in response to therapeutic interventions
Cancer biology:
Examining GCLC upregulation as a chemoresistance mechanism
Using GCLC as a biomarker for oxidative stress in tumor microenvironments
Targeting GCLC-dependent metabolic vulnerabilities in cancer cells
Cardiovascular conditions:
Metabolic disorders:
Exploring GCLC regulation in diabetic complications
Examining GCLC-dependent antioxidant responses in fatty liver disease
Investigating mitochondrial dysfunction through GCLC-dependent processes
Inflammatory conditions: