GLUL Antibody is employed in diverse assays to quantify or localize the enzyme:
Western Blot: Detects GLUL protein levels (e.g., in breast cancer cell lines or brain tissue) .
Immunohistochemistry (IHC): Identifies GLUL expression in astrocytes, liver cancer, or breast tumors .
Immunoprecipitation (IP): Isolates GLUL for downstream analysis (e.g., in HEK293 cells) .
Flow Cytometry: Measures intracellular GLUL in immune cells or cancer models .
GLUL is a key regulator of glutamine synthesis, a nutrient critical for cancer cell proliferation. Studies using GLUL Antibody reveal:
Breast Cancer: GLUL overexpression correlates with luminal subtype progression and tamoxifen resistance. GLUL knockdown reduces proliferation and colony formation in MCF7 cells .
Drug Resistance: GLUL downregulation in ALL or NSCLC cells (via siRNA or CRISPR) induces resistance to daunorubicin or docetaxel .
Immune Microenvironment: GLUL regulates IL-4 signaling, influencing immune cell growth and metabolism in breast cancer .
GLUL Antibody has facilitated novel metabolic studies:
Dual-Tracer SIRM: Combines ¹³C-glutamate and ¹⁵N-ammonium to trace GLUL activity. In HEK293 and HCT116 cells, GLUL-dependent glutamine synthesis supports nucleotide biosynthesis .
Inhibitor Studies: Methionine sulfoximine (MSO) blocks GLUL, halting proliferation in glutamine-dependent cancers .
GLUL is a marker for astrocytes, which detoxify neuronal glutamate by converting it to glutamine. Key findings include:
Neuroprotection: GLUL expression protects neurons from glutamate excitotoxicity .
Localization: IHC detects GLUL in astrocytes (e.g., mouse cerebellum) and neurons (e.g., cortical cultures) .
Glutamine synthetase (GLUL) is an enzyme responsible for converting glutamate and ammonia into glutamine. Glutamine plays a vital role as an energy source and participates in various cellular processes, including cell proliferation, apoptosis inhibition, and cell signaling. GLUL expression is observed during early fetal development and contributes to maintaining body pH by removing ammonia from circulation. Mutations in the GLUL gene have been linked to congenital glutamine deficiency.
The product appears as a colorless solution that has been sterilized through filtration.
The antibody solution has a concentration of 1mg/ml and is prepared in a buffer containing PBS with a pH of 7.4, 10% Glycerol, and 0.02% Sodium Azide.
For storage durations of up to one month, maintain the product at a temperature of 4°C. For extended storage periods, store at -20°C. It is important to avoid repeated freeze-thaw cycles.
The product remains stable for 12 months when stored at -20°C and for 1 month at 4°C.
The GLUL antibody has been rigorously tested using ELISA and Western blot analysis to ensure its specificity and reactivity. However, as applications may vary, it is recommended to perform reagent titration for each experiment to achieve optimal results.
GLNS, EC 6.3.1.2, EC 4.1.1.15, GLUL, Glutamine Synthetase, GS, Glutamate decarboxylase, Glutamate--ammonia ligase, PIG43, PIG59.
GLUL antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
PAT8D7AT.
Anti-human GLUL mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human GLUL amino acids 1-373 purified from E. coli.
Mouse IgG1 heavy chain and κ light chain.
GLUL (Glutamate-ammonia ligase) serves as the primary enzyme for ammonia detoxification and glutamate inactivation in the brain. It is predominantly expressed in astrocytes and catalyzes the conversion of glutamate to glutamine, playing a critical role in the glutamate-glutamine cycle. This enzyme is essential for neurotransmitter recycling and preventing excitotoxicity. Deficiency of GLUL has been associated with epilepsy and neurodegeneration, making it an important target for neuroscience research. Studies have demonstrated that selective deletion of GLUL in mouse cerebral cortex leads to progressive neurodegeneration and spontaneous seizures that increase in frequency with age . The enzyme's role in maintaining brain glutamate homeostasis positions it as a key focus for research into various neurological disorders.
GLUL antibodies are employed in various experimental applications including:
Immunohistochemistry and immunofluorescence for identifying astrocytes
Western blotting for quantifying GLUL protein expression
Flow cytometry for cell-specific analysis
Immunoprecipitation studies to investigate protein interactions
Validation of genetic knockout models such as the Emx1-Glul knockout mice described in the literature
These applications enable researchers to investigate the glutamate-glutamine cycle in normal and pathological brain states, examine astrocyte reactivity in disease models, and study cell-specific changes in GLUL expression under different experimental conditions.
GLUL antibodies provide valuable tools for investigating neurological disorders characterized by alterations in glutamate metabolism. Loss of astroglial GLUL has been reported in hippocampi of epileptic patients, and mouse models with GLUL deficiency exhibit reductions in tissue levels of aspartate, glutamate, glutamine, and GABA, along with decreased expression of glutamate receptor subunits and glutamate transporter proteins . Additionally, recent research has identified clustered de novo start-loss variants in GLUL that result in developmental and epileptic encephalopathy, characterized by drug-resistant epilepsy, global developmental delay, and hypotonia . By employing GLUL antibodies in these contexts, researchers can track changes in GLUL expression, localization, and post-translational modifications that may contribute to disease pathogenesis.
When working with GLUL antibodies, researchers should implement the following essential controls:
Genetic controls: Tissue from GLUL knockout or knockdown models (such as Emx1-Glul knockout mice)
Pre-absorption controls: Pre-incubating the antibody with purified GLUL protein
Multiple antibody validation: Using antibodies from different sources that recognize different epitopes
Western blot correlation: Confirming detection of a protein at the expected molecular weight
Primary antibody omission: To assess secondary antibody non-specific binding
Positive tissue controls: Liver or other tissues known to express high levels of GLUL
Housekeeping protein controls: For normalization in quantitative applications
These controls ensure specificity and reliability of GLUL antibody-based experiments, reducing the risk of false-positive or false-negative results.
Validating GLUL antibodies for specificity requires a multi-faceted approach:
Genetic validation: Use tissue from GLUL knockout models as negative controls. The selective deletion models described in the literature (like the Emx1-Glul knockout mice) provide excellent specificity controls .
Biochemical validation: Perform Western blotting to confirm a single band of appropriate molecular weight (~42 kDa). Multiple bands may indicate cross-reactivity or post-translational modifications.
Pre-absorption controls: Pre-incubate the antibody with purified GLUL protein before application to tissue. This should eliminate specific staining if the antibody is truly specific.
Multiple antibody approach: Use antibodies from different sources targeting different epitopes to confirm consistent staining patterns.
mRNA correlation: Compare antibody staining patterns with in situ hybridization or RNA sequencing data for GLUL expression.
Cross-species validation: If using the antibody across species, confirm specificity in each species separately.
Implementing these validation steps systematically ensures that experimental findings reflect true GLUL expression rather than antibody artifacts.
Different experimental applications require specific methodological considerations:
Sample preparation: Optimize protein extraction buffers to maintain GLUL integrity
Loading amount: Typically 10-30 μg total protein, depending on GLUL abundance
Blocking: 5% non-fat dry milk or 3-5% BSA in TBST
Primary antibody: Usually 1:1000 to 1:5000 dilution range
Secondary antibody: Typically 1:5000 to 1:20000
Detection system: Infrared detection systems (like Licor Odyssey) provide wider dynamic range for quantification
Fixation: 4% paraformaldehyde for 24-48 hours preserves GLUL antigenicity
Antigen retrieval: Often necessary, typically heat-induced in citrate buffer (pH 6.0)
Section thickness: 5-40 μm, depending on application
Primary antibody: 1:100 to 1:500 for paraffin sections; 1:200 to 1:1000 for frozen sections
Secondary antibody: 1:200 to 1:500
Controls: Include positive control tissue sections and primary antibody omission controls
Cell preparation: Gentle dissociation methods to preserve GLUL
Fixation/permeabilization: Critical for intracellular GLUL detection
Antibody concentration: Higher than for IHC, often 1:50 to 1:200
Controls: Include isotype controls and fluorescence minus one (FMO) controls
These methodological considerations must be optimized for each specific GLUL antibody and experimental system.
Recent research has identified start-loss variants in GLUL resulting in translation from downstream alternative start sites, producing truncated but enzymatically active GLUL proteins . Distinguishing between canonical GLUL and these variants requires strategic antibody selection:
Epitope-specific antibodies:
N-terminal antibodies: Will not detect truncated variants using alternative start sites
C-terminal antibodies: Will detect both canonical and truncated variants
Custom antibodies: May be designed to specifically target regions present or absent in variants
Combined techniques:
Western blotting: Canonical GLUL (~42 kDa) versus truncated variants (smaller size)
Immunoprecipitation followed by mass spectrometry: Can identify specific isoforms and post-translational modifications
2D gel electrophoresis: Can separate variants by both molecular weight and isoelectric point
Multiple antibody approach:
Use antibodies targeting different epitopes in parallel experiments
Compare results to infer the presence of specific variants
Genetic controls:
Express specific GLUL variants in cell systems as positive controls
Use CRISPR/Cas9 engineered cells expressing only specific variants
When studying start-loss variants like those described in developmental epileptic encephalopathy , researchers should carefully select antibodies that can differentiate between truncated and full-length forms.
Post-translational modifications (PTMs) of GLUL can significantly impact antibody detection:
N-terminal acetylation:
Research shows that glutamine levels regulate GLUL through an autoregulatory negative feedback mechanism involving N-terminal acetylation by p300/CREB binding protein
Antibodies targeting the N-terminus may show differential binding depending on acetylation status
This modification is particularly relevant when studying the regulatory mechanisms of GLUL
Ubiquitination:
Phosphorylation:
GLUL activity can be modulated by phosphorylation
Phospho-specific antibodies may be needed to detect activated forms
Standard GLUL antibodies may show altered binding to phosphorylated forms
Experimental considerations:
Include phosphatase or deubiquitinase treatments to assess PTM contributions
Use multiple antibodies targeting different epitopes
Consider native versus denaturing conditions for detection
Understanding these PTM effects is crucial when studying GLUL regulation, particularly in pathological conditions where regulatory mechanisms may be disrupted.
Tissue preparation significantly impacts GLUL antibody detection:
Fixation protocols:
4% paraformaldehyde (PFA) fixation for 24-48 hours typically preserves GLUL antigenicity while maintaining good tissue morphology
Avoid overfixation which can mask epitopes through excessive cross-linking
Post-fixation in 30% sucrose before freezing helps preserve tissue integrity for cryosectioning
Section preparation:
For immunofluorescence: 10-40 μm sections are optimal (thinner for high-resolution imaging, thicker for 3D analysis)
For chromogenic IHC: 5-10 μm sections provide better resolution
Free-floating sections often provide better antibody access than slide-mounted sections
Antigen retrieval methods:
Heat-induced epitope retrieval (HIER) in citrate buffer (pH 6.0) often improves GLUL detection
Microwave, water bath, or pressure cooker methods may yield different results
Proteolytic epitope retrieval is generally less effective for GLUL
Storage considerations:
Fixed tissue: Store at 4°C in PBS with 0.02% sodium azide for short-term or in cryoprotectant for long-term
Frozen sections: Store at -80°C and use within 6-12 months
Paraffin blocks: Can be stored at room temperature for years without significant antigen loss
These preparation methods should be systematically optimized for each specific GLUL antibody to achieve consistent and reliable results.
Optimal antibody conditions vary by application and must be determined empirically:
Application | Primary Antibody Range | Secondary Antibody Range | Incubation Conditions | Key Considerations |
---|---|---|---|---|
Western blot | 1:1000-1:5000 | 1:5000-1:20000 | 4°C overnight or 1-2 hours at RT | Total protein amount, detection method sensitivity |
IHC (paraffin) | 1:100-1:500 | 1:200-1:500 | 4°C overnight | Antigen retrieval method, section thickness |
IHC (frozen) | 1:200-1:1000 | 1:200-1:500 | 4°C overnight | Fixation duration, detergent concentration |
Immunofluorescence | 1:100-1:500 | 1:200-1:500 | 4°C overnight | Background autofluorescence, multiplexing considerations |
Flow cytometry | 1:50-1:200 | N/A (often direct conjugates) | 30-60 minutes at RT or 4°C | Cell permeabilization method, fixation impact |
For all applications, researchers should:
Perform antibody titration experiments to determine optimal concentration
Consider increased antibody concentration for fixed tissues versus fresh/frozen
Adjust incubation time inversely with concentration (more dilute solutions may require longer incubation)
Optimize blocking conditions to improve signal-to-noise ratio
Consider the use of signal amplification systems for low-abundance detection
These parameters should be systematically tested and documented for reproducible GLUL detection.
Common issues with GLUL antibody experiments and their solutions include:
High background/non-specific binding:
Increase blocking time and concentration (try 5-10% normal serum)
Add 0.1-0.3% Triton X-100 or Tween-20 to reduce hydrophobic interactions
Use more dilute antibody solutions
Increase wash steps in number and duration
Try different secondary antibodies with minimal species cross-reactivity
For fluorescence applications, include anti-autofluorescence treatments
Weak or absent signal:
Optimize antigen retrieval (try different pH buffers and heating methods)
Reduce fixation time or switch fixation method
Increase antibody concentration or incubation time
Try signal amplification systems (e.g., tyramide signal amplification)
Confirm sample preparation preserves GLUL (avoid excessive protease treatments)
Test antibody on known positive control tissue
Inconsistent staining across sections:
Ensure uniform section thickness
Process all sections simultaneously in the same reagents
Use adequate volume of antibody solution to cover sections completely
Avoid tissue folding or overlapping
Implement humidity chambers to prevent edge drying
Western blot issues:
Multiple bands: Check for protein degradation, splice variants, or post-translational modifications
No bands: Verify protein extraction method preserves GLUL, check transfer efficiency
Smeared bands: Reduce protein loading, optimize gel percentage, check for protein aggregation
Systematic troubleshooting with appropriate controls at each step can resolve most GLUL antibody detection issues.
Quantification of GLUL expression changes requires rigorous methodology:
Western blotting quantification:
Perform antibody dilution series to ensure detection in the linear range
Include recombinant GLUL protein standards for absolute quantification
Normalize to multiple housekeeping proteins or total protein staining
Use infrared detection systems for wider dynamic range and better quantification
Analyze multiple biological replicates (n≥3) for statistical validity
Immunohistochemical quantification:
Use stereological approaches rather than simple cell counting
Analyze multiple brain regions and multiple sections per animal
Standardize image acquisition parameters (exposure time, gain settings)
Conduct analysis blinded to experimental condition
Consider automated image analysis algorithms to reduce bias
Flow cytometry quantification:
Use calibration beads to standardize fluorescence intensity
Report mean fluorescence intensity (MFI) rather than percent positive
Include isotype controls for background correction
Consider cell type-specific markers for subpopulation analysis
Data normalization strategies:
For tissue with altered astrocyte populations: normalize to astrocyte-specific markers
For developmental studies: age-matched controls are essential
For disease models: include both positive and negative controls
When comparing across experiments: include internal reference standards
Statistical considerations:
Use appropriate statistical tests based on data distribution
Account for multiple comparisons when analyzing multiple brain regions
Consider region-specific and cell type-specific changes separately
Report effect sizes in addition to p-values
These quantification approaches ensure reliable measurement of GLUL expression changes in various experimental and disease contexts.
Multiplexed imaging with GLUL antibodies enables complex analyses of astrocyte function in relation to other cell types and proteins:
Antibody selection for multiplexing:
Choose GLUL antibodies raised in different host species than other target antibodies
Verify that secondary antibodies have minimal cross-reactivity
Consider directly conjugated primary antibodies to reduce species limitations
Test for epitope masking or steric hindrance when targeting multiple proteins
Technical approaches:
Spectral unmixing: Use spectral imaging systems to separate overlapping fluorophores
Sequential staining: Apply, image, and strip or quench antibodies in sequence
Tyramide signal amplification: Allows use of multiple primary antibodies from the same species
Expansion microscopy: Physically expands tissue to improve resolution of co-localized proteins
Analysis methods:
Co-localization analysis: Quantify overlap between GLUL and other markers
Spatial relationship analysis: Measure distances between GLUL+ cells and other cell types
Morphometric analysis: Quantify astrocyte morphology in relation to GLUL expression levels
Cell-specific expression profiling: Correlate GLUL levels with other astrocyte markers
Application examples:
Multiplexed imaging approaches provide rich contextual information about GLUL function within complex neural circuits.
Recent research has revealed a glutamine-dependent feedback mechanism regulating GLUL expression, where elevated glutamine triggers GLUL degradation through N-terminal modification . Researchers can study this mechanism using:
Antibody-based approaches:
N-terminal specific antibodies to detect acetylation status
Ubiquitin co-immunoprecipitation to assess ubiquitination of GLUL
Comparison of antibodies targeting different epitopes to assess structural changes
Pulse-chase experiments combined with immunoprecipitation to measure GLUL turnover
Experimental systems:
Analytical methods:
Western blotting to assess total GLUL levels under different glutamine conditions
Immunofluorescence to examine subcellular localization changes
FRET/FLIM approaches to study protein-protein interactions in the degradation pathway
Mass spectrometry to identify post-translational modifications
Genetic approaches:
Compare wild-type GLUL with N-terminal variants resistant to acetylation
Study regulation in cells with knocked-down p300/CREB binding protein or CRL4CRBN
Use CRISPR/Cas9 to introduce specific mutations in the N-terminal regulatory region
These approaches can help elucidate how disruption of this autoregulatory feedback mechanism contributes to the developmental consequences observed in patients with GLUL start-loss variants .
Emerging methodologies are expanding the capabilities of GLUL antibody research:
Super-resolution microscopy techniques:
STED, STORM, and PALM imaging overcome diffraction limits
Allows visualization of GLUL distribution within astrocyte microdomains
Enables co-localization studies at nanometer resolution
In vivo antibody-based imaging:
Antibody-based PET ligands for non-invasive GLUL imaging
Cleared tissue approaches (CLARITY, iDISCO) for whole-brain GLUL mapping
In vivo labeling of surface-expressed proteins combined with post-mortem GLUL detection
Single-cell proteomics:
Mass cytometry (CyTOF) for high-dimensional analysis of GLUL in relation to dozens of other proteins
Single-cell Western blotting for heterogeneity assessment
Spatial proteomics techniques to preserve regional information
Functional antibody approaches:
Function-blocking antibodies to inhibit GLUL activity in specific compartments
Activity-based GLUL sensors using antibody fragments
Intrabodies expressed in specific cell populations to track GLUL dynamics
Automated high-throughput approaches:
Tissue microarrays for rapid screening across multiple conditions
Machine learning algorithms for automated GLUL quantification
High-content screening platforms to identify regulators of GLUL expression
These emerging methodologies offer unprecedented insights into GLUL biology and its role in neurological disorders.
GLUL antibody research has significant potential for therapeutic development in neurological disorders:
Diagnostic applications:
Development of antibody-based assays to detect altered GLUL levels in cerebrospinal fluid
Immunohistochemical assessment of GLUL in surgical specimens from epilepsy patients
Identification of patient subgroups with specific GLUL alterations who might respond to targeted therapies
Target validation:
Use of GLUL antibodies to validate mouse models of GLUL-related disorders
Confirmation of drug effects on GLUL expression and activity
Correlation of GLUL changes with clinical outcomes
Therapeutic monitoring:
Assessment of treatment effects on GLUL expression and localization
Monitoring of compensatory changes in glutamate/glutamine metabolism
Evaluation of astrocyte responses to therapeutic interventions
Drug development applications:
Precision medicine approaches:
Stratification of patients based on GLUL expression patterns
Identification of individuals likely to benefit from glutamine/glutamate metabolism-targeted therapies
Development of personalized therapeutic strategies for patients with specific GLUL variants
GLUL antibody research provides critical tools for understanding pathological mechanisms and developing targeted interventions for epilepsy and developmental disorders associated with glutamine metabolism dysfunction.
Mouse anti-human antibodies are monoclonal antibodies produced in mice that are specific for human antigens. These antibodies are widely used in research, diagnostics, and therapeutic applications due to their high specificity and affinity for their target antigens .
The production of mouse anti-human antibodies involves immunizing mice with a specific human antigen, followed by the isolation of B cells that produce antibodies against the antigen. These B cells are then fused with myeloma cells to create hybridomas, which can be cultured to produce large quantities of monoclonal antibodies .
Mouse anti-human antibodies are used in various applications, including:
One of the challenges associated with the use of mouse anti-human antibodies in therapeutic applications is the potential for the human immune system to recognize these antibodies as foreign and mount an immune response against them. This response, known as the Human Anti-Mouse Antibody (HAMA) response, can reduce the efficacy of the treatment and cause adverse effects .
To mitigate the HAMA response, various strategies have been developed, including the humanization of mouse antibodies. This involves modifying the antibody’s structure to increase the proportion of human sequences while retaining the specificity and affinity for the target antigen .