GSS Antibody

Glutathione Synthetase, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Definition and Types of GSS Antibodies

GSS antibodies are immunoglobulins (IgG) that specifically bind to glutathione synthetase, an enzyme that catalyzes the ATP-dependent condensation of γ-glutamylcysteine and glycine to form glutathione . These antibodies are classified into two main types:

  • Monoclonal Antibodies: Derived from hybridomas (e.g., mouse anti-human GSS clones like PAT12F2AT ).

  • Polyclonal Antibodies: Produced by immunizing animals (e.g., rabbits) with recombinant GSS or synthetic peptides .

Key differences include specificity (monoclonal vs. broad polyclonal) and host species (mouse, rabbit, or others).

Applications in Research

GSS antibodies are employed in diverse experimental settings:

Western Blot (WB)

Used to quantify GSS protein levels in tissues (e.g., liver, brain) and cell lines (e.g., HEK-293, CHO) . For example:

  • Dilution: 1:1,000–1:4,000 .

  • Controls: Positive signals in human liver, mouse brain, and kidney .

Immunohistochemistry (IHC)

Detects GSS localization in human kidney (with antigen retrieval) and neuronal tissues . For instance, GSS antibodies revealed dopaminergic neurodegeneration in Gerstmann–Sträussler–Scheinker disease (GSS P102L) .

Immunofluorescence (IF/ICC)

Visualizes subcellular distribution (e.g., cytoplasmic GSS in MDCK cells) .

ELISA

Measures GSS activity or serum levels, aiding in studies of oxidative stress .

Inflammation and Oxidative Stress

  • Anti-inflammatory Effects: GSS antibodies demonstrated that glutathione synthetase inhibits NF-κB activation and reduces TNF-α/IL-6 secretion in lipopolysaccharide (LPS)-stimulated endothelial cells, mitigating sepsis-induced acute lung injury .

  • Oxidative Damage: In Gss−/− mice, GSS deficiency led to glutathione depletion, elevated ROS levels, and testicular ferroptosis due to lipid peroxidation and autophagy dysregulation .

Disease Pathogenesis

  • Genetic Deficiencies: GSS mutations cause 5-oxoprolinuria, characterized by metabolic acidosis, hemolytic anemia, and CNS dysfunction .

  • Neurodegeneration: Prion protein deposition in GSS P102L mutants correlates with nigrostriatal dopamine system degeneration, detected via GSS antibodies in post-mortem brain samples .

Therapeutic Implications

GSS antibodies are critical in validating GSS-targeting therapies. For example:

  • KO Validation: Assay Genie’s CAB14535 antibody confirmed GSS knockout efficacy in cellular models .

  • Biomarker Studies: ELISA-based GSS quantification aids in monitoring glutathione metabolism in cancer, neurodegenerative diseases, and metabolic disorders .

Challenges and Considerations

FactorRecommendationSource
Cross-reactivityVerify species specificity (e.g., human vs. rodent) Proteintech, Bioss
StorageStore at -20°C; avoid freeze-thaw cycles Prospec, Proteintech
DilutionOptimize per assay (e.g., WB: 1:1,000–1:4,000) Proteintech, Bioss

Product Specs

Introduction
Glutathione synthetase (GSS) is a crucial enzyme in the glutathione biosynthesis pathway. It catalyzes the final step, where gamma-glutamylcysteine and glycine combine to form glutathione. Deficiencies in GSS lead to a condition called glutathione synthetase deficiency, also known as GSS deficiency, 5-oxoprolinuria, or pyroglutamic aciduria. This condition is characterized by increased hemolysis (destruction of red blood cells) and impaired central nervous system function.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The antibody is supplied as a 1 mg/ml solution in a buffer consisting of phosphate-buffered saline (PBS) at a pH of 7.4, 10% glycerol, and 0.02% sodium azide.
Storage Procedures
For short-term storage (up to 1 month), keep the antibody at 4°C. For long-term storage, store at -20°C. Repeated freezing and thawing should be avoided.
Stability / Shelf Life
The antibody is stable for 12 months when stored at -20°C and for 1 month at 4°C.
Applications
This GSS antibody has been validated for use in various applications, including ELISA, Western blot analysis, and immunocytochemistry/immunofluorescence (ICC/IF), ensuring its specificity and reactivity. However, it is recommended to optimize the antibody concentration for each specific application to achieve the best results.
Synonyms
Glutathione synthetase, GSH synthetase, GSH-S, Glutathione synthase, GSHS, MGC14098, GSS.
Purification Method
GSS antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT12F2AT.
Immunogen
Anti-human GSS mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human GSS amino acids 1-474 purified from E. coli.
Ig Subclass
Mouse IgG1 heavy chain and k light chain.

Q&A

What is GSS and what is its biological significance?

Glutathione synthetase (GSS) is a critical enzyme involved in the synthesis of glutathione, an essential antioxidant that protects cells from oxidative stress and maintains cellular homeostasis. GSS plays a crucial role in various cellular processes including signal transduction and cell proliferation. Its biological significance extends to multiple pathways that regulate oxidative balance, which is vital for normal cellular function .

GSS has a calculated molecular weight of 52 kDa (474 amino acids) and is encoded by the GSS gene (NCBI Gene ID: 2937). The protein is highly conserved across species, showing reactivity in human, mouse, rat, pig, canine, and hamster samples, indicating its evolutionary importance in cellular metabolism .

What applications are GSS antibodies commonly used for in research?

GSS antibodies are versatile tools in molecular and cellular research with multiple validated applications:

ApplicationCommon Uses
Western Blot (WB)Protein expression analysis, molecular weight confirmation
Immunohistochemistry (IHC)Tissue localization studies, expression pattern analysis
Immunofluorescence (IF)/ICCSubcellular localization, co-localization studies
ELISAQuantitative protein measurement

These applications enable researchers to investigate GSS expression patterns, localization, and potential alterations in various physiological and pathological conditions . Each application requires specific optimization for optimal results, and published literature demonstrates successful use across these methodologies .

How does GSS antibody validation differ from other antibody validations?

GSS antibody validation involves several specific considerations due to the enzyme's role in glutathione metabolism. Unlike antibodies against structural proteins or receptors, GSS antibody validation requires confirmation of specificity against an enzyme with catalytic activity.

A robust validation approach includes knockout (KO) validation, where the antibody shows no signal in GSS knockout samples, confirming specificity. This is particularly important given that metabolic enzymes may have structural homology with related proteins in the same pathway . Cross-reactivity testing against related enzymes in the glutathione metabolism pathway is essential to ensure specific targeting of GSS without detecting glutathione peroxidase or glutathione reductase.

Additionally, validation across multiple species is important for comparative studies, as GSS shows high conservation but may have species-specific epitopes that affect antibody recognition .

How should I design experiments to investigate GSS expression in disease models?

When investigating GSS expression in disease models, a comprehensive experimental design should incorporate multiple approaches:

First, establish baseline GSS expression in your model system using Western blot analysis with recommended dilutions (1:1000-1:4000) of the GSS antibody . Include appropriate positive controls (liver tissue samples are good positive controls as they express high levels of GSS) and negative controls.

For tissue studies, immunohistochemistry with carefully optimized antigen retrieval conditions is critical. The search results indicate that TE buffer pH 9.0 is suggested for GSS antibody (15712-1-AP), though citrate buffer pH 6.0 may also be used . This optimization is crucial as improper antigen retrieval can lead to false negative results.

For cellular localization studies, immunofluorescence at 1:50-1:500 dilution can reveal subcellular distribution patterns of GSS . Compare distribution between normal and disease models to identify potential alterations in localization.

When working with multiple species, be aware that antibody reactivity may vary. Validate the antibody in each species before conducting comparative studies. The GSS antibody mentioned in the search results shows reactivity with human, mouse, rat, pig, canine, and hamster samples .

Finally, incorporate functional assays that correlate GSS protein levels with glutathione synthesis capacity to establish biological relevance of any observed changes in expression.

How can I optimize GSS antibody use for detecting low-abundance expression?

Detecting low-abundance GSS expression requires methodological optimization across several parameters:

Signal Amplification Techniques: For Western blot, consider using enhanced chemiluminescence (ECL) substrates with higher sensitivity or fluorescence-based detection systems. Longer exposure times may be necessary, but be cautious of increased background.

Sample Enrichment: For tissues or cells with low GSS expression, consider subcellular fractionation to concentrate the cytosolic fraction where GSS is predominantly localized. This can increase the signal-to-noise ratio significantly.

Antibody Concentration Optimization: While standard dilutions are recommended (1:1000-1:4000 for WB) , for low-abundance samples, higher concentrations may be necessary. Conduct a titration series to determine the optimal antibody concentration that maximizes specific signal while minimizing background.

Blocking Optimization: Test different blocking agents (BSA, non-fat milk, commercial blockers) to reduce background that might obscure weak signals. For the GSS antibody, optimizing blocking conditions can significantly improve detection sensitivity.

Signal Development Time: For enzymatic detection methods, optimize the signal development time. Extended development can enhance detection of low-abundance proteins but may increase background.

Tissue-specific Considerations: For IHC detection of low GSS expression, consider using amplification systems such as tyramide signal amplification after validating specificity with conventional methods.

What are the critical considerations when using GSS antibodies for evaluating oxidative stress responses?

When using GSS antibodies to evaluate oxidative stress responses, several critical factors must be considered:

Temporal Dynamics: GSS expression changes in response to oxidative stress follow specific time courses. Design time-course experiments to capture both acute (minutes to hours) and chronic (days) responses. Cell type-specific responses may vary significantly, so pilot studies are essential.

Co-analysis with Other Oxidative Stress Markers: GSS changes should be interpreted alongside other glutathione metabolism enzymes and oxidative stress markers. Consider parallel assessment of glutathione peroxidase, glutathione reductase, and reduced/oxidized glutathione ratios for comprehensive pathway analysis.

Specificity Controls: Include appropriate controls to distinguish GSS-specific responses from general stress responses. This might include treatments that induce other stress pathways without directly affecting oxidative balance.

Functional Correlation: Correlate GSS protein levels with enzymatic activity assays to determine whether increased protein expression translates to enhanced glutathione synthesis. This is particularly important as post-translational modifications of GSS may affect its activity independently of expression levels.

Subcellular Redistribution: Oxidative stress may trigger subcellular redistribution of GSS without changing total protein levels. Use immunofluorescence or subcellular fractionation followed by Western blotting to detect such changes .

Degradation Products: Be aware that oxidative stress can increase protein degradation, resulting in antibody-reactive fragments. Use Western blotting to verify that the detected signal corresponds to the expected molecular weight of GSS (52 kDa) .

What are the optimal protocols for different GSS antibody applications?

Each application of GSS antibody requires specific optimization for reliable results:

Western Blot Protocol Optimization:

  • Use recommended dilutions (1:1000-1:4000)

  • Sample preparation: Standard protein extraction with protease inhibitors

  • Loading: 20-40 μg of total protein per lane

  • Transfer: Standard wet transfer protocols work well for GSS (52 kDa)

  • Blocking: 5% non-fat milk in TBST for 1 hour at room temperature

  • Primary antibody incubation: Overnight at 4°C in blocking buffer

  • Secondary antibody: Anti-rabbit IgG-HRP at 1:5000 dilution

  • Detection: Standard ECL methods with exposure times adjusted for signal strength

Immunohistochemistry Protocol Optimization:

  • Recommended dilution: 1:100-1:400

  • Fixation: 10% neutral buffered formalin

  • Antigen retrieval: TE buffer pH 9.0 (primary recommendation) or citrate buffer pH 6.0 (alternative)

  • Blocking: 10% normal serum in PBS for 1 hour

  • Primary antibody incubation: Overnight at 4°C

  • Detection system: HRP-polymer based systems

  • Counterstain: Hematoxylin for nuclear visualization

Immunofluorescence Protocol Optimization:

  • Recommended dilution: 1:50-1:500

  • Fixation: 4% paraformaldehyde for 15 minutes

  • Permeabilization: 0.2% Triton X-100 for 10 minutes

  • Blocking: 3% BSA in PBS for 30 minutes

  • Primary antibody incubation: 2 hours at room temperature or overnight at 4°C

  • Secondary antibody: Fluorophore-conjugated anti-rabbit at 1:500

  • Counterstain: DAPI for nuclear visualization

These protocols should be further optimized for specific sample types and research questions. Always include appropriate positive controls (liver tissue for WB, kidney tissue for IHC) and negative controls in each experiment.

How can I troubleshoot inconsistent GSS antibody results?

Inconsistent GSS antibody results can stem from various factors. Here's a systematic approach to troubleshooting:

No Signal or Weak Signal:

  • Antibody concentration: Increase antibody concentration or incubation time

  • Antigen retrieval (for IHC): Test both recommended methods - TE buffer pH 9.0 and citrate buffer pH 6.0

  • Protein degradation: Ensure sample integrity with fresh protease inhibitors

  • Detection system: Check secondary antibody reactivity and detection reagent activity

  • Exposure time: Increase for Western blot applications

High Background:

  • Blocking conditions: Increase blocking time or agent concentration

  • Antibody concentration: Reduce primary antibody concentration

  • Washing: Increase wash duration and number of wash steps

  • Secondary antibody: Reduce concentration or switch to more specific alternatives

  • Endogenous peroxidase (for IHC): Ensure proper quenching

Multiple Bands in Western Blot:

  • Sample degradation: Use fresher samples with protease inhibitors

  • Non-specific binding: Optimize blocking and washing conditions

  • Cross-reactivity: Validate with GSS knockout controls

  • Post-translational modifications: Consider that additional bands might represent modified GSS forms

  • Expected molecular weight: Confirm against reference (52 kDa for GSS)

Inconsistent Staining Patterns in IHC/IF:

  • Fixation time: Standardize fixation protocols

  • Antigen retrieval: Ensure consistent heating and buffer conditions

  • Section thickness: Maintain uniform section thickness

  • Antibody penetration: Adjust permeabilization conditions

  • Cell/tissue heterogeneity: Consider biological variability in GSS expression

Document all optimization steps systematically to establish a reliable protocol for your specific experimental system.

How can I quantify GSS expression levels accurately using antibody-based methods?

Accurate quantification of GSS expression using antibody-based methods requires careful attention to methodology:

Western Blot Quantification:

  • Loading controls: Use housekeeping proteins (β-actin, GAPDH) or total protein staining methods (Ponceau S)

  • Linear dynamic range: Perform dilution series to establish linear detection range

  • Multiple exposures: Capture both weak and strong signals without saturation

  • Normalization: Normalize GSS signal to loading controls

  • Replication: Perform at least three biological replicates

  • Software analysis: Use specialized image analysis software that accounts for background

Immunohistochemistry Quantification:

  • Standardized staining: Process all samples simultaneously

  • Digital image analysis: Use consistent acquisition settings

  • Region selection: Analyze multiple fields per sample (minimum 5-10)

  • Scoring systems: Develop clear intensity and distribution scoring criteria

  • Automated analysis: Consider using machine learning-based image analysis for objectivity

  • Controls: Include positive and negative controls in each batch

ELISA-based Quantification:
While specific information on GSS ELISA wasn't provided in the search results, general principles apply:

  • Standard curve: Generate with recombinant GSS protein

  • Sample dilution: Test multiple dilutions to ensure measurements within linear range

  • Technical replicates: Minimum of duplicate measurements

  • Spike recovery: Validate accuracy with known protein addition

  • Inter-assay calibration: Include common samples across multiple plates

For all quantification methods, statistical validation of reproducibility is essential. Consider using coefficient of variation calculations to assess method precision and reliability.

How do I interpret changes in GSS expression in relation to disease mechanisms?

Interpreting changes in GSS expression in disease contexts requires a multi-faceted approach:

Baseline Context: Establish normal GSS expression patterns in relevant tissues and cell types. Expression can vary significantly between tissues, with liver and kidney showing relatively high expression levels .

Magnitude Assessment: Consider both statistical and biological significance of observed changes. Small but statistically significant changes may not have biological impact, while large fold-changes warrant further investigation.

Pathway Integration: Interpret GSS changes within the broader glutathione synthesis pathway. Parallel assessment of γ-glutamylcysteine synthetase and glutathione levels provides context for GSS alterations.

Causal vs. Compensatory Changes: Determine whether GSS alterations are driving pathology or represent compensatory responses. Temporal studies can help distinguish between these possibilities.

Cellular Specificity: In heterogeneous tissues, identify specific cell types showing altered GSS expression using co-localization studies with cell-type markers.

Functional Consequences: Correlate GSS expression changes with glutathione levels and oxidative stress markers to establish functional impact.

Disease-Specific Considerations:

  • In cancer: Increased GSS may indicate adaptation to elevated oxidative stress and potential chemoresistance

  • In neurodegenerative diseases: Decreased GSS might contribute to oxidative vulnerability

  • In metabolic disorders: Alterations may reflect disrupted redox homeostasis

Translational Relevance: Consider how GSS changes might inform therapeutic strategies, such as targeting glutathione metabolism or addressing oxidative stress.

How can GSS antibodies be used in multi-parameter analysis of oxidative stress responses?

GSS antibodies can be integrated into comprehensive multi-parameter analyses of oxidative stress pathways:

Multiplexed Immunofluorescence Approaches:
Combine GSS antibody with antibodies against other glutathione pathway proteins and oxidative stress markers. Design panels including:

  • GSS (glutathione synthesis)

  • Glutathione peroxidase (glutathione utilization)

  • Glutathione reductase (glutathione recycling)

  • Nrf2 (master regulator of antioxidant response)

  • 4-HNE or 8-OHdG (oxidative damage markers)

Ensure antibody compatibility by selecting those raised in different host species or using directly conjugated primary antibodies.

Sequential Immunoblotting:
For Western blot applications, sequential probing or multiplex fluorescent detection can be used to analyze multiple proteins from the same samples. This approach preserves precious samples and reduces inter-blot variability.

Flow Cytometry Integration:
For cellular studies, GSS antibody can be incorporated into flow cytometry panels alongside:

  • Mitochondrial ROS indicators (MitoSOX)

  • Glutathione content measurements (monochlorobimane)

  • Cell death markers (Annexin V/PI)

  • Cell type-specific markers

Single-Cell Approaches:
Combine GSS immunostaining with single-cell transcriptomics to correlate protein expression with gene expression patterns at the single-cell level, revealing population heterogeneity in oxidative stress responses.

Spatial Analysis in Tissues:
Use multiplexed IHC or immunofluorescence to map GSS expression in relation to tissue microenvironment features, including hypoxic regions, inflammatory infiltrates, or proliferative zones.

Integration with Functional Assays:
Correlate GSS immunostaining with functional measurements such as glutathione content, reactive oxygen species levels, or cell viability under oxidative challenge.

What are the limitations of GSS antibody-based research that researchers should be aware of?

Researchers should be cognizant of several limitations when using GSS antibodies:

Post-translational Modification Detection:
Standard GSS antibodies may not distinguish between various post-translationally modified forms of the protein, which could have different activities. Phosphorylation, ubiquitination, or other modifications may alter GSS function without changing detectable levels.

Correlation with Activity:
Antibody detection quantifies protein presence but does not directly measure enzymatic activity. GSS enzyme activity assays should complement antibody-based detection for functional studies.

Sensitivity Limitations:
Detection thresholds may miss low-level expression in certain cell types or conditions. This limitation is particularly relevant in tissues with naturally low GSS expression.

Temporal Resolution:
Antibody-based methods provide snapshots of expression but cannot capture real-time dynamics of GSS regulation. Complementary approaches like reporter systems may be needed for dynamic studies.

Fixation and Processing Artifacts:
For histological applications, fixation and processing can affect epitope accessibility and produce artifacts. Validation across multiple fixation protocols is advisable for novel applications.

Quantification Challenges:
Semi-quantitative methods like Western blot have inherent limitations in accuracy and precision. Standardization with recombinant protein controls and robust normalization strategies can mitigate but not eliminate these challenges.

Isoform Detection:
Depending on the epitope targeted, antibodies may not detect all splice variants or isoforms of GSS, potentially missing biologically relevant forms of the protein.

How can machine learning approaches enhance GSS antibody data analysis?

Machine learning approaches offer powerful tools for enhancing GSS antibody data analysis across multiple dimensions:

Automated Image Analysis for IHC/IF:
Deep learning models can improve quantification of GSS staining patterns in tissues and cells, offering advantages including:

  • Consistent intensity scoring across large datasets

  • Cell-type specific expression quantification in heterogeneous tissues

  • Subcellular localization pattern recognition

  • Correlation with tissue morphological features

  • Multiplexed staining analysis with reduced observer bias

Predicting Antibody Binding Profiles:
As demonstrated in search result , machine learning models can predict antibody binding properties, which could inform:

  • Selection of optimal GSS antibody clones for specific applications

  • Design of new antibodies with enhanced specificity for GSS

  • Prediction of cross-reactivity with related proteins

  • Optimization of binding conditions for challenging applications

Integration of Multi-omics Data:
Machine learning can integrate GSS antibody-derived data with:

  • Transcriptomic profiles to correlate protein and mRNA levels

  • Metabolomic data to connect GSS levels with glutathione metabolism

  • Clinical parameters to identify biomarker potential

Predictive Modeling of GSS in Disease:
Machine learning models trained on GSS expression data could:

  • Identify patterns associated with disease progression

  • Predict response to therapies targeting oxidative stress

  • Classify patient subgroups based on GSS-related parameters

Experimental Design Optimization:
Models can recommend optimal experimental conditions for GSS antibody applications:

  • Antibody dilution optimization based on sample characteristics

  • Protocol parameters for specific tissue types

  • Troubleshooting recommendations for failed experiments

As noted in search result , large antibody datasets with quantitative binding measurements provide essential training data for these machine learning approaches, enabling increasingly sophisticated analysis tools for GSS research.

What is the potential for using GSS antibodies in multiplex assays for systems biology research?

GSS antibodies hold significant potential for integration into multiplex assays that support systems biology research:

Multiplexed Protein Analysis Platforms:
Technologies such as:

  • Multiplex immunofluorescence

  • Mass cytometry (CyTOF)

  • Digital spatial profiling

  • Microarray-based antibody platforms

These allow simultaneous detection of GSS alongside dozens to hundreds of other proteins, enabling comprehensive pathway analysis of glutathione metabolism within broader cellular networks.

Single-Cell Proteomics Integration:
GSS antibody incorporation into single-cell proteomics workflows can reveal:

  • Cell-to-cell variability in antioxidant defense mechanisms

  • Correlation between GSS expression and cellular phenotypes

  • Identification of previously unrecognized cell subpopulations based on stress response profiles

Spatial Transcriptomics Correlation:
Combining GSS immunostaining with spatial transcriptomics can:

  • Map protein expression against transcriptional landscapes

  • Identify post-transcriptional regulation of GSS

  • Characterize microenvironmental influences on GSS expression

Pathway Activation Mapping:
Including GSS in phospho-protein panels can connect glutathione metabolism to:

  • Stress-activated protein kinase pathways

  • NRF2-KEAP1 signaling cascades

  • Cell survival and death decision pathways

Biomarker Panel Development:
GSS antibodies can contribute to multiplex biomarker panels for:

  • Oxidative stress assessment in clinical samples

  • Drug response prediction

  • Disease progression monitoring

Organoid and 3D Culture Applications:
Multiplex imaging including GSS can characterize:

  • Zonal patterns of antioxidant capacity in complex cultures

  • Dynamic responses to oxidative challenges in physiologically relevant models

For optimal integration into multiplex assays, GSS antibodies need thorough validation for specificity, sensitivity, and compatibility with multiplex protocols, including appropriate epitope retrieval methods as specified in the search results .

How can GSS antibodies contribute to neurodegenerative disease research?

GSS antibodies offer valuable tools for investigating oxidative stress mechanisms in neurodegenerative diseases:

Regional Vulnerability Assessment:
GSS immunohistochemistry can map differential expression across brain regions, potentially explaining selective vulnerability to neurodegenerative processes. Using dilutions of 1:100-1:400 as recommended , researchers can quantify region-specific variations in GSS expression that may correlate with disease-affected areas.

Cellular Stress Response Profiling:
Combining GSS antibodies with neuronal, astrocytic, and microglial markers using multiplex immunofluorescence can reveal cell type-specific alterations in glutathione metabolism. The recommended 1:50-1:500 dilution range for immunofluorescence provides flexibility for optimization in neuronal tissues.

Disease Progression Monitoring:
GSS antibody-based assays can track temporal changes in glutathione synthesis capacity across disease stages in both animal models and human postmortem samples. Western blotting at 1:1000-1:4000 dilution enables quantitative assessment of GSS protein levels throughout disease progression.

Therapeutic Response Assessment:
For experimental treatments targeting oxidative stress, GSS antibodies can evaluate intervention effects on glutathione synthesis capacity. This is particularly valuable for compounds designed to upregulate antioxidant defense mechanisms.

Genetic Model Validation:
In transgenic models of neurodegenerative diseases, GSS antibodies can confirm downstream effects of genetic modifications on antioxidant defense systems. The knockout-validated antibodies are especially valuable for distinguishing specific from non-specific effects.

Protein Aggregation Studies:
Co-localization studies using GSS antibodies alongside antibodies against disease-specific protein aggregates (Aβ, tau, α-synuclein) can investigate relationships between defective glutathione metabolism and protein misfolding.

Blood-Brain Barrier Analysis:
GSS immunostaining can assess vascular contributions to oxidative vulnerability by examining GSS expression in blood-brain barrier components under disease conditions.

These applications provide mechanistic insights into oxidative stress contributions to neurodegeneration and potential therapeutic targets for intervention.

What are the best practices for using GSS antibodies in cancer research applications?

Cancer research applications of GSS antibodies require specific considerations for optimal results:

Tumor Heterogeneity Analysis:
Cancer tissues often show significant heterogeneity in antioxidant capacity. When using GSS antibodies for IHC (at 1:100-1:400 dilution) , analyze multiple regions within tumors to capture this heterogeneity. Quantify both staining intensity and percentage of positive cells across representative fields.

Therapy Resistance Mechanisms:
For studies examining chemotherapy or radiation resistance:

  • Compare GSS expression before and after treatment

  • Correlate GSS levels with treatment response

  • Combine with other drug resistance markers

Western blotting at 1:1000-1:4000 dilution provides quantitative comparison between sensitive and resistant cell lines or tissues.

Metabolic Reprogramming Assessment:
Cancer cells undergo metabolic reprogramming that includes altered redox balance. Use GSS antibodies in conjunction with markers of metabolic pathways (glycolysis, glutaminolysis) to establish connections between energy metabolism and redox control.

Stromal Interaction Studies:
For tumor microenvironment research:

  • Use dual immunofluorescence with GSS (1:50-1:500) and stromal markers

  • Assess GSS expression in cancer-associated fibroblasts, tumor-associated macrophages, and endothelial cells

  • Investigate paracrine regulation of glutathione metabolism

Patient-Derived Xenograft Applications:
When working with PDX models:

  • Validate GSS antibody reactivity with both human (tumor) and mouse (stroma) GSS

  • Be aware of species cross-reactivity as indicated in the antibody specifications

  • Use species-specific secondary antibodies for differentiation

Experimental Controls:
Include appropriate controls:

  • High GSS-expressing tissues (liver, kidney) as positive controls

  • GSS-knockdown cancer cells as negative controls

  • Normal adjacent tissue for baseline comparison

Prognostic/Predictive Biomarker Development:
For biomarker studies:

  • Standardize staining protocols (antigen retrieval with TE buffer pH 9.0)

  • Establish clear scoring criteria

  • Correlate with clinical outcomes in adequately powered cohorts

Following these practices will enhance the reliability and translational relevance of GSS antibody applications in cancer research.

How can cutting-edge technologies enhance GSS antibody specificity and applications?

Recent technological advances offer opportunities to enhance GSS antibody specificity and expand application capabilities:

Recombinant Antibody Technology:
Moving beyond traditional polyclonal antibodies like those described in the search results , recombinant antibody technologies enable:

  • Precise epitope targeting for enhanced GSS specificity

  • Elimination of batch-to-batch variability

  • Engineering of detection tags and functional modifications

  • Development of single-chain variable fragments (scFvs) for specialized applications

Machine Learning-Guided Antibody Design:
As highlighted in search results and , machine learning approaches can:

  • Predict optimal antibody sequences for GSS binding

  • Design GSS-specific binding profiles with minimal cross-reactivity

  • Optimize antibody stability and performance characteristics

  • Select ideal CDR modifications for enhanced sensitivity

Nanobody and Alternative Scaffold Technologies:
These emerging technologies offer advantages including:

  • Smaller size for improved tissue penetration in GSS detection

  • Enhanced stability for challenging applications

  • Reduced immunogenicity for in vivo applications

  • Capacity for site-specific conjugation

Super-Resolution Microscopy Compatibility:
Next-generation GSS antibodies optimized for:

  • STORM/PALM microscopy for nanoscale GSS localization

  • Expansion microscopy compatibility

  • Correlative light and electron microscopy applications

  • Multiplexed super-resolution imaging

In Situ Proximity Ligation Approaches:
These techniques can:

  • Detect GSS interactions with other proteins in the glutathione synthesis pathway

  • Visualize post-translational modifications of GSS

  • Provide enhanced sensitivity for low-abundance GSS detection

Mass Spectrometry Immunoassays:
Combining GSS antibodies with mass spectrometry enables:

  • Detection of specific GSS proteoforms

  • Absolute quantification of GSS protein levels

  • Identification of post-translational modifications

  • Multiplexed analysis with other proteins

These technological advances significantly expand the research capabilities for GSS investigation beyond the traditional applications documented in the search results , enabling more sophisticated analyses of GSS in complex biological systems.

What developments in quantitative serology assays could improve GSS antibody research?

Advances in quantitative serology assays offer substantial opportunities for enhancing GSS antibody research precision and applications:

Reference Standard Development:
As described in search result for virus antibody quantification, development of reference standards for GSS antibody assays would enable:

  • Standardized quantitation across laboratories

  • Absolute protein quantification rather than relative measurements

  • Enhanced reproducibility and comparability between studies

  • Calibration of diverse assay platforms

Digital ELISA Technologies:
Emerging ultrasensitive detection platforms provide:

  • Single-molecule detection capability for low-abundance GSS

  • Expanded dynamic range spanning several orders of magnitude

  • Reduced sample volume requirements

  • Improved precision at low concentrations

Automated Liquid Handling Integration:
Robotics-assisted assay platforms enable:

  • Higher throughput for population-scale GSS studies

  • Improved precision through elimination of manual variability

  • Standardized processing across large sample sets

  • Integrated quality control measures

Multiplexed Bead-Based Assays:
Technologies such as Luminex offer:

  • Simultaneous quantification of GSS alongside other glutathione pathway proteins

  • Reduced sample consumption for multi-parameter analysis

  • Internal controls within each assay

  • Flexible assay design for project-specific requirements

Microfluidic and Point-of-Care Applications:
Miniaturized systems provide advantages including:

  • Rapid GSS quantification in time-sensitive applications

  • Minimal reagent consumption

  • Potential for field application in environmental studies

  • Integration with other metabolic measurements

Enhanced Data Analysis Algorithms:
Computational approaches improve quantification through:

  • Automated curve-fitting optimized for GSS concentration ranges

  • Outlier detection and management

  • Plate effect normalization

  • Integration with experimental metadata for comprehensive analysis

These developments parallel the approaches outlined for virus antibody quantification in search result , where reference detection antibodies standardized quantitation across diverse samples. Similar principles applied to GSS quantification would significantly advance research capabilities in this field.

What are the essential controls for GSS antibody experiments?

Implementing comprehensive controls is critical for reliable GSS antibody experiments:

Positive Controls:

  • Tissue-specific positive controls: Liver and kidney tissues show reliable GSS expression and serve as excellent positive controls for antibody validation

  • Cell line controls: HEK-293 and CHO cells have been validated for GSS expression and can serve as reliable positive controls

  • Recombinant GSS protein: When available, purified protein provides definitive size control for Western blot applications

Negative Controls:

  • Antibody omission controls: Primary antibody omission identifies non-specific binding of secondary antibodies

  • Isotype controls: Non-specific rabbit IgG at matching concentration to rule out host-specific background

  • Knockdown/knockout controls: Cells with GSS gene silencing or knockout provide the gold standard for antibody specificity validation

Procedural Controls:

  • Antigen competition: Pre-incubation of antibody with purified GSS protein should eliminate specific staining

  • Cross-reactivity assessment: Testing against related enzymes in the glutathione pathway

  • Dilution series: Serial antibody dilutions to establish optimal signal-to-noise ratio

  • Multiple detection methods: Confirmation of findings using alternative antibody-based techniques

Quantification Controls:

  • Loading controls for Western blot: Housekeeping proteins or total protein staining

  • Internal reference standards: Calibrated recombinant protein for quantitative applications

  • Batch controls: Common samples processed across multiple experimental runs to assess inter-assay variability

Biological Context Controls:

  • Physiological modulation: Samples with expected GSS upregulation (e.g., oxidative stress induction) or downregulation

  • Species-specific controls: When working across species, validation in each species of interest

  • Developmental stage controls: When relevant, age-matched controls for developmental studies

Implementing these controls ensures experimental validity and facilitates troubleshooting when unexpected results occur.

How should researchers approach GSS antibody selection for specific research questions?

Strategic antibody selection is crucial for addressing specific GSS research questions effectively:

Research Question-Driven Selection:
Begin by clearly defining your research objective:

  • Protein localization studies require antibodies validated for IHC/IF (dilution 1:100-1:400 for IHC, 1:50-1:500 for IF)

  • Quantitative expression analysis needs antibodies optimized for Western blot (dilution 1:1000-1:4000)

  • Protein interaction studies benefit from antibodies targeting non-interaction domains

  • Functional studies require antibodies that don't interfere with enzymatic activity

Technical Considerations:

  • Host species: Consider compatibility with other antibodies in multiplex applications

  • Clonality: Polyclonal antibodies like those in the search results offer high sensitivity but may have batch variability; monoclonal antibodies provide consistency

  • Format: Primary antibodies versus conjugated formats for direct detection

  • Size: Full IgG versus fragments (Fab, F(ab')2) for specific applications

Validation Documentation:
Assess available validation data:

  • Knockout validation provides the highest specificity confidence

  • Multiple application validation demonstrates versatility

  • Publication history indicates successful use in peer-reviewed research

  • Species cross-reactivity documentation for comparative studies

Epitope Considerations:

  • Target region: N-terminal, C-terminal, or internal epitopes may be differentially accessible

  • Post-translational modifications: Some epitopes may be masked by phosphorylation or other modifications

  • Conformational sensitivity: Some antibodies recognize only native or denatured GSS

Practical Factors:

  • Application-specific optimization data

  • Stability and storage requirements

  • Lot-to-lot consistency

  • Cost-benefit analysis for project needs

Experimental Validation:
Plan preliminary validation in your specific experimental system:

  • Pilot testing in representative samples

  • Comparison of multiple antibodies when possible

  • Optimization of protocols for your specific application

By systematically evaluating these factors, researchers can select GSS antibodies that best match their specific research questions and experimental systems.

What are the best practices for reporting GSS antibody-based research results?

Adhering to rigorous reporting standards ensures research reproducibility and reliability:

Antibody Identification Details:

  • Complete antibody identification: Catalog number, clone/lot number, manufacturer

  • RRID (Research Resource Identifier) inclusion when available (e.g., AB_2878171 for GSS antibody)

  • Host species and clonality (e.g., Rabbit polyclonal)

  • Immunogen information when available (e.g., GSS fusion protein Ag8338)

Experimental Methodology:

  • Detailed protocol description including buffer compositions

  • Antibody dilutions used for each application (e.g., 1:1000-1:4000 for WB)

  • Antigen retrieval methods for IHC (e.g., TE buffer pH 9.0 or citrate buffer pH 6.0)

  • Detection systems and imaging parameters

  • Software used for image analysis and quantification

Validation Information:

  • Positive and negative controls employed

  • Validation experiments conducted

  • Known limitations or caveats

  • Cross-reactivity assessment

  • Knockout/knockdown validation if performed

Results Presentation:

  • Representative images showing entire field of view

  • Unmodified primary data with scale bars

  • Quantification methods clearly described

  • Statistical analysis details

  • Raw data availability statement

Reproducibility Considerations:

  • Sample sizes and biological replicates

  • Technical replicate information

  • Batch effects management

  • Inter-observer validation for subjective assessments

Citation of Related Methods:

  • Previous validation studies

  • Original antibody characterization

  • Methodological precedents

This comprehensive reporting aligns with the ARRIVE guidelines for animal research and enhances experimental reproducibility across laboratories. Standardized reporting facilitates meta-analysis and systematic reviews of GSS-related research.

What emerging questions about GSS could be addressed using advanced antibody-based approaches?

Several cutting-edge research questions about GSS can be addressed using advanced antibody-based approaches:

Spatial Proteomics of GSS in Cellular Stress Responses:
Using highly multiplexed antibody imaging technologies, researchers could map the dynamic redistribution of GSS in relation to other oxidative stress response proteins during various stress conditions. This would reveal spatial coordination of antioxidant defense mechanisms at subcellular resolution.

GSS Post-Translational Modification Landscape:
Development of modification-specific antibodies could uncover how post-translational modifications regulate GSS activity and stability. This approach could identify novel regulatory mechanisms affecting glutathione synthesis capacity in health and disease.

Cell-Type Specific GSS Regulation in Complex Tissues:
Advanced multiplex immunofluorescence combined with spatial transcriptomics could reveal cell type-specific GSS expression patterns and regulatory mechanisms in heterogeneous tissues, particularly in the brain and immune system.

GSS Protein-Protein Interaction Networks:
Proximity ligation assays using GSS antibodies could map the dynamic interactome of GSS under various cellular conditions, potentially uncovering novel regulatory proteins and signaling pathways that modulate glutathione synthesis.

Single-Cell GSS Variability in Disease Progression:
Single-cell antibody-based technologies could reveal how GSS expression heterogeneity within cell populations contributes to differential vulnerability to disease processes, particularly in cancer and neurodegeneration.

Circadian Regulation of GSS Expression:
Temporal analysis of GSS expression and localization over circadian cycles could uncover time-dependent regulation of antioxidant capacity, with implications for chronopharmacology and optimal timing of interventions targeting oxidative stress.

GSS in Extracellular Vesicles and Intercellular Communication:
Investigating GSS packaging into extracellular vesicles using antibody-based approaches could reveal novel mechanisms of intercellular communication in redox regulation.

Developmental Trajectory of GSS Expression: Tracking GSS expression through developmental stages could identify critical periods where glutathione synthesis capacity is established, with implications for developmental origins of disease susceptibility.

Product Science Overview

Introduction

Glutathione synthetase (GSS) is an essential enzyme in the biosynthesis of glutathione, a critical antioxidant that protects cells from oxidative stress and maintains redox balance. The enzyme catalyzes the ATP-dependent conversion of γ-glutamylcysteine and glycine to glutathione. Glutathione synthetase is encoded by the GSS gene and is highly conserved across species, including humans and mice.

Mouse Anti-Human Glutathione Synthetase Antibody

Mouse anti-human glutathione synthetase antibodies are monoclonal antibodies produced by immunizing mice with human GSS protein. These antibodies are designed to specifically bind to human GSS, allowing researchers to detect and quantify the enzyme in various biological samples. The antibodies are commonly used in immunohistochemistry (IHC), Western blotting (WB), and enzyme-linked immunosorbent assays (ELISA).

Preparation Methods

The preparation of mouse anti-human glutathione synthetase antibodies involves several key steps:

  1. Immunization: Mice are immunized with recombinant human GSS protein to elicit an immune response.
  2. Hybridoma Production: Spleen cells from the immunized mice are fused with myeloma cells to create hybridoma cells that produce monoclonal antibodies.
  3. Screening and Selection: Hybridoma cells are screened for the production of antibodies that specifically bind to human GSS. Positive clones are selected and expanded.
  4. Purification: The monoclonal antibodies are purified from the hybridoma cell culture supernatant using protein A or protein G affinity chromatography.
Applications

Mouse anti-human glutathione synthetase antibodies have several important applications in biomedical research:

  • Immunohistochemistry (IHC): These antibodies are used to detect GSS expression in tissue sections, providing insights into the enzyme’s distribution and localization in different tissues.
  • Western Blotting (WB): The antibodies are employed to detect and quantify GSS protein levels in cell lysates and tissue extracts, aiding in the study of protein expression and regulation.
  • ELISA: These antibodies are utilized in ELISA assays to measure GSS concentrations in biological samples, facilitating research on glutathione metabolism and related diseases.

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.