GSLs are cell membrane components composed of ceramide and glycan chains. Their oligosaccharide structures vary by cell lineage, differentiation stage, and pathology, enabling their use as markers for immune targeting .
Antigenicity: GSLs with C24 ceramide chains exhibit strong immunogenicity, inducing IgG3-class antibodies via thymus-independent type 2 (TI-2) mechanisms .
Applications:
| GSL Feature | Antibody Class | Mechanism | Reference |
|---|---|---|---|
| C24 ceramide | IgG3 | TI-2 antigen-like activation | |
| Oligosaccharide structure | Oligoclonal | Glycan-specific epitope binding |
GLS-010 is a clinical-stage IgG4 monoclonal antibody targeting PD-1, a key immune checkpoint in cancer immunotherapy .
Blocks PD-1 interaction with PD-L1/L2, reactivating T-cell responses .
Exhibits high affinity (EC50 ~210 pM for human PD-1) and specificity, avoiding cross-reactivity with CD28/CTLA-4 family proteins .
Efficacy:
Safety:
| Parameter | Result | Reference |
|---|---|---|
| Objective Response Rate | 23.6% | |
| Complete Responses | 10 patients | |
| Median Progression-Free Survival | Not reported |
This rabbit polyclonal antibody targets glutaminase (GLS), a mitochondrial enzyme critical for glutamine metabolism .
Research: Used in immunofluorescence (IF), immunohistochemistry (IHC), and western blotting (WB) to study GLS expression in cancer cells .
Specifications:
GLS (Glutaminase) is an enzyme that catalyzes the first reaction in the primary pathway for renal catabolism of glutamine, converting glutamine to glutamate. This conversion supports the tricarboxylic acid cycle and redox and epigenetic reactions . GLS antibodies are critical research tools because:
They enable detection and quantification of GLS expression in various tissues and cell types
They help investigate the role of glutaminolysis in cancer metabolism and other diseases
They support studies into the metabolic reprogramming of cancer cells
They facilitate research into neurological disorders, as GLS regulates levels of glutamate, the main excitatory neurotransmitter in the brain
GLS has multiple isoforms produced by alternative splicing that researchers should consider when selecting antibodies:
| Isoform | Alternative Names | Molecular Weight | Characteristics |
|---|---|---|---|
| KGA | Kidney-type glutaminase | ~73 kDa (calculated) | Primary isoform in kidney |
| GAC | Glutaminase C | ~58-65 kDa (observed) | Common in cancer cells |
| GAM | Glutaminase M | ~63 kDa | Found in various tissues |
Some antibodies like Proteintech's 23549-1-AP can recognize all three isoforms (KGA, GAM, GAC) of GLS , which is an important consideration when designing experiments to study specific or multiple isoforms.
Optimizing GLS antibody performance in Western blotting requires attention to several technical details:
Expected molecular weight considerations: GLS has a calculated molecular weight of 73 kDa, but observed bands may appear at 58-65 kDa depending on the isoform . This discrepancy is normal and has been validated in published studies.
Sample preparation protocol:
Use fresh tissue/cells and maintain cold conditions throughout
Include protease inhibitors in lysis buffer to prevent degradation
Consider phosphatase inhibitors as GLS activity can be regulated by phosphorylation
For mitochondrial proteins like GLS, ensure proper subcellular fractionation if studying specific compartments
Antibody validation strategy:
Recommended blocking conditions: 5% non-fat milk in TBST is typically effective, though specific antibodies may have optimized conditions in their datasheets .
To ensure reproducibility in immunohistochemistry with GLS antibodies:
Antigen retrieval optimization: For GLS antibodies, suggested antigen retrieval includes:
Fixation considerations:
Signal amplification methods:
For low-expressing samples, consider tyramide signal amplification
Biotin-based amplification systems should be used cautiously as endogenous biotin can create background in metabolically active tissues
Validation across multiple tissues:
When selecting a GLS antibody for specific research applications, consider:
Epitope location relative to functional domains:
Cross-reactivity with species of interest:
Validation data availability:
Ability to distinguish between isoforms:
Some antibodies recognize all GLS isoforms
Others may be isoform-specific
Select based on whether your research questions require isoform discrimination
GLS antibodies are instrumental in studying cancer metabolism:
Comparative expression analysis techniques:
Co-localization studies:
Immunofluorescence using GLS antibodies can reveal subcellular localization changes in cancer cells
Combined with mitochondrial markers, researchers can study altered compartmentalization
Patient sample stratification:
Experimental models:
GLS antibodies can help investigate the complex relationship between GLS expression and the tumor immune microenvironment:
Correlation with immune cell infiltration:
High GLS expression correlates with altered immune cell infiltration patterns:
Multi-parameter immunofluorescence techniques:
Combined staining with GLS antibodies and immune cell markers can reveal spatial relationships
This approach can map metabolic zones within tumors and their impact on immune cell function
Mechanistic studies of metabolic immune suppression:
Immunotherapy response prediction:
High GLS expression is associated with lower MSI, TMB, and neoantigen count in multiple cancer types
This correlates with potentially weaker response to immune checkpoint inhibitor therapy
In COAD, STAD, UCEC, SKCM, and OV, high GLS expression predicts poorer response to various immunotherapeutic approaches
GLS antibodies are essential tools for evaluating glutaminase inhibitor efficacy:
Target engagement assessment:
Western blotting with GLS antibodies can confirm that inhibitors are reaching their target
Changes in post-translational modifications can be monitored using phospho-specific antibodies
Pharmacodynamic marker development:
IHC staining of tumor biopsies pre- and post-treatment can measure GLS levels as a pharmacodynamic marker
Combined with downstream metabolite measurements, this creates a comprehensive view of drug activity
Resistance mechanism investigation:
In resistant models, GLS antibodies can help identify changes in expression, localization, or isoform switching
Immunoprecipitation followed by mass spectrometry can identify novel binding partners in resistant contexts
Combination therapy rationale development:
IHC co-staining of GLS with other metabolic enzymes can identify patients who might benefit from combination approaches
For example, tumors with both high GLS and high PD-L1 might benefit from combined glutaminase inhibitors and immune checkpoint inhibitors
Research has revealed critical roles for GLS in T cell biology that can be studied using GLS antibodies:
GLS knockout and inhibition studies:
GLS-deficient T cells show increased IFNγ expression in Th1-skewing conditions
Conversely, they show decreased IL17A expression in Th17-skewing conditions
This differential effect is linked to transcription factor expression patterns, with increased T-bet in Th1 and decreased RORγt in Th17 conditions
Metabolic flux analysis:
Temporal analysis of metabolic adaptation:
During early activation (days 1-2), both Th1 and Th17 cells show reduced transcription factors and cell size with GLS inhibition
By day 5, Th1 cells recover and show increased cell size and T-bet expression
Total rRNA levels reflect these changes, with similar levels at day 3 but divergent patterns by day 5
Methodology considerations:
Use flow cytometry with GLS antibodies to quantify GLS levels at single-cell resolution during differentiation
Combine with metabolic dyes to correlate GLS expression with functional metabolic states
Consider subcellular fractionation to track potential redistribution during activation
Flow cytometry with GLS antibodies presents several technical challenges:
Intracellular staining optimization:
GLS is primarily located in mitochondria, requiring effective permeabilization
Standard paraformaldehyde fixation followed by saponin or Triton X-100 permeabilization works for most applications
For co-staining with surface markers, use a sequential staining approach: surface markers first, followed by fixation, permeabilization, and GLS antibody staining
Signal-to-noise considerations:
Autofluorescence can be a challenge in metabolically active cells with high mitochondrial content
Use appropriate fluorochromes that excite/emit away from cellular autofluorescence peaks
Include FMO (fluorescence minus one) controls to set accurate gates
Epitope preservation:
Some fixation methods may mask GLS epitopes
Test multiple permeabilization reagents (e.g., Triton X-100, saponin, methanol) to determine optimal conditions
Consider using indirect staining with secondary antibodies for signal amplification
Validation strategies:
Use GLS knockout or knockdown cells as negative controls
Compare expression patterns with known GLS expression data across immune cell types
Confirm flow cytometry results with other methods (e.g., Western blot, qPCR)
The relationship between GLS expression and immunotherapy response can be investigated through several approaches:
Multiplex immunohistochemistry:
Combine GLS antibodies with immune checkpoint markers (PD-1, PD-L1, CTLA-4)
Quantify spatial relationships between GLS-high tumor regions and immune infiltrates
This approach reveals the metabolic landscape that may affect immunotherapy efficacy
Correlation with immunotherapy response biomarkers:
Immunophenotyping studies:
GLS expression correlates with specific changes in tumor immune microenvironment:
Negative correlation with CD8+ T lymphocyte abundance in HNSC, UCEC, and CESC
Positive correlation with cancer-associated fibroblast infiltration across almost all tumors
Positive correlation with immunosuppressive MDSC infiltration in COAD, HNSC, LGG, LIHC, and UCEC
Combination therapy research models:
For tumors with high GLS expression, combining glutaminase inhibitors with immune checkpoint inhibitors may overcome resistance
Mouse models with humanized immune systems can be used to test such combinations
GLS antibodies are essential tools to monitor on-target effects in these models
Inconsistent molecular weight observations with GLS antibodies can be addressed methodically:
Understanding expected weight variations:
Post-translational modification considerations:
Phosphorylation of GLS can alter its mobility on SDS-PAGE
Proteolytic processing may generate fragments
Try phosphatase treatment of samples to determine if modifications affect migration
Sample preparation adjustments:
Use freshly prepared samples when possible
Include multiple protease inhibitors in lysis buffer
Avoid freeze-thaw cycles that may lead to degradation
Consider non-reducing conditions if disulfide bonds affect epitope recognition
Gel system optimization:
Use gradient gels (4-20%) to better resolve proteins across a wide molecular weight range
Adjust running conditions (voltage, time) to improve resolution in the relevant size range
Consider native PAGE if protein folding affects antibody recognition
Non-specific staining with GLS antibodies can be addressed through several approaches:
Blocking optimization:
Test different blocking solutions (BSA, normal serum, commercial blockers)
Extend blocking time to reduce background
Use the blocking solution as antibody diluent to maintain blocking during incubation
Antibody validation and specificity:
Verify antibody specificity through knockout/knockdown controls
Peptide competition assays can confirm specificity
Pre-adsorption of the antibody with recombinant antigen can reduce non-specific binding
Sample-specific challenges:
Some tissues (e.g., liver, kidney) may have high endogenous biotin, causing background with biotin-based detection systems
Endogenous peroxidase activity in tissues like spleen can cause background with HRP-based systems
Use appropriate quenching steps (hydrogen peroxide treatment for peroxidase, avidin/biotin blocking for biotin)
Protocol modifications:
Increase washing steps in number and duration
Add detergent (0.1-0.3% Triton X-100) to reduce hydrophobic interactions
Titrate primary antibody to find optimal concentration balancing signal and background
For immunofluorescence, include an autofluorescence quenching step
When faced with contradictory results from different GLS antibodies:
Epitope mapping and comparison:
Validation through orthogonal methods:
Confirm protein expression using mRNA analysis (qPCR, RNA-seq)
Use mass spectrometry-based proteomics for unbiased protein detection
Apply genetic approaches (siRNA, CRISPR) to validate antibody specificity
Isoform-specific considerations:
Determine if contradictory results stem from isoform-specific detection
Use isoform-specific primers in qPCR to correlate with protein findings
Consider that different isoforms may predominate in different tissues or conditions
Documentation and reporting practices:
Thoroughly document all experimental conditions
Report the specific antibody clone, catalog number, and lot in publications
Include detailed methods sections describing validation approaches
Contact antibody manufacturers with contradictory findings to contribute to knowledge base
Combining GLS antibodies with single-cell technologies enables powerful analyses of metabolic heterogeneity:
Single-cell Western blotting approaches:
Microfluidic platforms can perform Western blots on individual cells
GLS antibodies can quantify expression at single-cell resolution
This reveals population heterogeneity masked in bulk analyses
Mass cytometry (CyTOF) applications:
Metal-conjugated GLS antibodies can be incorporated into CyTOF panels
Combined with functional markers, this approach can correlate GLS expression with cell state
Allows simultaneous measurement of up to 40 parameters, including multiple metabolic enzymes
Spatial transcriptomics correlation:
GLS antibody staining on serial sections can be correlated with spatial transcriptomics data
This approach maps metabolic zones within tumors or tissues
Reveals relationships between GLS expression and local microenvironmental features
Methodological considerations:
Validate antibodies specifically for single-cell applications
Include appropriate single-cell controls
Consider fixation and permeabilization conditions that preserve both target epitopes and cellular morphology
Developing companion diagnostics with GLS antibodies requires specific considerations:
Standardization requirements:
Rigorous antibody validation across multiple laboratories
Establishment of scoring systems for quantitative assessment
Development of reference standards for calibration
Predictive biomarker selection:
Determine whether total GLS protein levels, specific isoforms, or post-translational modifications best predict response
Correlate IHC findings with functional metabolic measurements
Develop multi-parameter predictive algorithms that may include GLS along with other markers
Technical standardization for clinical implementation:
Automated staining platforms to ensure reproducibility
Digital pathology approaches for objective quantification
Standard operating procedures that can be implemented across clinical laboratories
Validation in clinical trial contexts:
Retrospective analysis of samples from glutaminase inhibitor trials
Prospective collection in basket trials to correlate expression with response
Establishment of clinically meaningful cutoffs for "high" versus "low" expression
Investigating metabolic enzyme networks using GLS antibodies involves several approaches:
Co-immunoprecipitation studies:
Use GLS antibodies for immunoprecipitation followed by mass spectrometry
Identify novel interaction partners in different cellular contexts
Validate findings with reciprocal co-IPs using antibodies against identified partners
Multiplex immunofluorescence techniques:
Combine GLS antibodies with antibodies against other metabolic enzymes
This approach reveals spatial relationships and potential metabolic compartmentalization
Quantify co-localization using digital image analysis
Proximity ligation assay (PLA):
Use PLA to detect proteins in close proximity (<40 nm)
This technique can reveal transient or weak interactions between GLS and other proteins
Provides spatial resolution beyond conventional co-localization studies
Chromatin immunoprecipitation (ChIP) studies:
Investigate whether GLS or its metabolic products affect transcriptional regulation
Use antibodies against histone modifications affected by glutamine metabolism
Correlate findings with GLS expression and activity levels