G6PD Antibody

Glucose-6-Phosphate Dehydrogenase, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Definition and Types of G6PD Antibodies

G6PD antibodies are designed to target the G6PD enzyme, which catalyzes the first step of the pentose phosphate pathway, generating NADPH to protect cells from oxidative damage. Two primary types are available:

TypeSourceApplicationsKey Vendors
MonoclonalMouse/RabbitWestern Blot (WB), IHC, IP, ICC/IFR&D Systems (MAB11467), Cell Signaling (CST #8866)
PolyclonalRabbitWB, IHC, IP, ELISA, CoIPProteintech (25413-1-AP), Abcam (ab993)

Key Features:

  • Specificity: Detects G6PD at ~59 kDa (reducing conditions) .

  • Reactivity: Primarily human, with cross-reactivity to mouse, rat, and sheep in some cases .

  • Validation: Tested in WB, IHC, IP, and ICC/IF .

Applications in Research

G6PD antibodies are employed across diverse fields, including cancer biology, immunology, and genetic disorders.

Immunogenic Cell Death and Cancer Therapy

Inhibition of G6PD induces immunogenic cell death (ICD) in tumors, enhancing immunotherapy efficacy. Studies using G6PD antibodies demonstrated:

  • Mechanism: G6PD knockdown reduces NADPH, increasing oxidative stress and triggering ICD markers (e.g., calreticulin translocation, HMGB1 release) .

  • Therapeutic Synergy: Combining G6PD inhibition with immune checkpoint inhibitors (ICIs) improved tumor control in melanoma and lung cancer models .

Autoimmune and Infectious Diseases

G6PD deficiency is linked to heightened autoimmune susceptibility. Antibodies aid in studying:

  • Autoimmune Markers: Elevated antinuclear (OR 1.81), anti-Smith (OR 5.00), and anti-RNP-68 (OR 4.00) antibodies in G6PD-deficient cohorts .

  • Phagocytic Dysfunction: Reduced oxidative burst in severe G6PD deficiency, impairing bacterial clearance .

Diagnostic and Biomarker Research

G6PD antibodies enable precise quantification of enzyme levels:

  • Serological Testing: Detection of G6PD activity in erythrocytes and leukocytes .

  • Cancer Prognosis: Low G6PD expression correlates with better outcomes in melanoma and lung cancer patients treated with ICIs .

Clinical Relevance

G6PD antibodies bridge basic research and clinical practice:

  • Drug Development: Screening for G6PD inhibitors that synergize with ICIs in oncology .

  • Personalized Medicine: Identifying G6PD-deficient patients at risk of hemolysis or autoimmune flares .

Emerging Trends

  • Biomarker Discovery: Profiling G6PD expression in autoimmune diseases (e.g., lupus, rheumatoid arthritis) .

  • Infectious Disease: Studying G6PD’s role in sepsis and mycobacterial susceptibility .

Product Specs

Introduction
Glucose-6-phosphate dehydrogenase (G6PD) is an essential enzyme in the pentose phosphate pathway, responsible for producing NADPH. This molecule is crucial for maintaining cellular redox balance by reducing oxidative stress. G6PD deficiency can lead to hemolytic anemia, neonatal jaundice, and acute hemolysis due to the impaired ability to produce NADPH and protect red blood cells from damage.
Formulation
The antibody is supplied at a concentration of 1mg/ml in a solution of phosphate-buffered saline (PBS) at pH 7.4, with 0.1% sodium azide as a preservative.
Storage Procedures
For short-term storage (up to 1 month), keep the antibody at 4°C. For longer-term storage, it is recommended to store at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
The antibody is stable for 12 months when stored at -20°C and for 1 month when stored at 4°C.
Synonyms
G6PD, G6PD1, Glucose-6-phosphate 1-dehydrogenase.
Purification Method
G6PD antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT2F6AT.
Immunogen
Anti-human G6PD mAb is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human G6PD amino acids 35-506 purified from E. coli.
Ig Subclass
Mouse IgG2b heavy chain and κ light chain.

Q&A

What is G6PD and what role does it play in cellular metabolism?

G6PD (Glucose-6-phosphate dehydrogenase) is an essential enzyme that catalyzes the conversion of glucose-6-phosphate to 6-phosphogluconolactone while reducing NADP+ to NADPH in the pentose phosphate pathway. This enzyme plays multiple critical roles including: maintaining NADPH levels which protects cells from oxidative damage, supporting red blood cell function, providing pentoses for nucleotide synthesis, and contributing to cellular redox homeostasis .

G6PD deficiency is a genetic disorder that is inherited from parent to child and can cause destruction of red blood cells leading to hemolytic anemia. This condition predominantly affects men, though women can be carriers of the defective gene . The deficiency is more common in people of African, Mediterranean, or Asian descent and typically manifests when individuals are exposed to triggers such as fava beans, certain medications, or infections .

In cancer biology, G6PD has gained significant attention as it is frequently upregulated in various cancer types, contributing to enhanced metabolism, increased stress tolerance, and resistance to therapeutic interventions .

How do G6PD antibodies function in research applications?

G6PD antibodies are specialized immunoglobulins designed to recognize and bind specifically to the G6PD protein. In research settings, they function through highly specific epitope recognition that enables multiple analytical applications. These antibodies can be monoclonal (derived from a single B-cell clone, offering high specificity) or polyclonal (derived from multiple B-cell lineages, providing stronger signals by recognizing multiple epitopes).

The functional principles of G6PD antibodies in research include:

  • Specific binding to G6PD protein epitopes with high affinity

  • Enabling detection through secondary detection systems (such as HRP-conjugated secondary antibodies)

  • Facilitating visualization and quantification of G6PD expression in various biological samples

  • Supporting analysis of G6PD's subcellular localization and potential interaction partners

When properly validated, G6PD antibodies provide researchers with powerful tools to investigate the expression, regulation, and function of this enzyme across diverse biological systems and disease states .

What are the most common applications for G6PD antibodies in academic research?

G6PD antibodies are employed across multiple research methodologies, with each providing distinct advantages for investigating different aspects of G6PD biology:

Western Blot Analysis:

  • Detection of G6PD protein in cell lysates, showing characteristic bands at approximately 58-59 kDa

  • Quantitative comparison of G6PD expression across different experimental conditions

  • Evaluation of G6PD levels between normal and pathological samples

Immunohistochemistry (IHC):

  • Visualization of G6PD expression patterns in tissue sections

  • Assessment of subcellular localization (primarily cytoplasmic)

  • Correlation of expression patterns with histopathological features

Simple Western™ (Automated Capillary-Based Western Blotting):

  • More quantitative and reproducible analysis of G6PD protein levels

  • Higher throughput analysis across multiple samples

Flow Cytometry:

  • Single-cell analysis of G6PD expression

  • Correlation with other cellular markers in heterogeneous populations

Immunoprecipitation:

  • Investigation of G6PD protein-protein interactions

  • Analysis of post-translational modifications affecting G6PD activity

Each application requires specific optimization and validation to ensure reliable and reproducible results, with Western blot and IHC being the most commonly documented methods in current G6PD research literature.

What are the optimal protocols for using G6PD antibodies in Western blot analysis?

Achieving optimal results with G6PD antibodies in Western blot analysis requires careful attention to sample preparation, protein separation, and detection protocols. Based on established methodologies, the following protocol offers reliable G6PD detection:

Sample Preparation:

  • Lyse cells in an appropriate buffer containing protease inhibitors

  • Quantify protein concentration using Bradford or BCA assays

  • Prepare samples under reducing conditions with loading buffer containing SDS and β-mercaptoethanol

Gel Electrophoresis and Transfer:

  • Load 15-30 μg of protein per lane on a 10-12% SDS-PAGE gel

  • Include validated positive control samples (e.g., A549 or MCF-7 cell lysates)

  • Transfer proteins to PVDF membrane using standard transfer conditions

Antibody Incubation:

  • Block membrane with 5% non-fat dry milk or BSA in TBST for 1 hour at room temperature

  • Incubate with anti-G6PD primary antibody at 1 μg/ml concentration overnight at 4°C

  • Wash membrane thoroughly with TBST (3-5 washes, 5 minutes each)

  • Incubate with appropriate HRP-conjugated secondary antibody (e.g., anti-mouse IgG for monoclonal antibodies)

  • Wash thoroughly to remove unbound secondary antibody

Detection:

  • Apply ECL substrate and detect signal via chemiluminescence

  • G6PD should appear as a specific band at approximately 58-59 kDa

  • For quantitative analysis, normalize to appropriate loading controls

When troubleshooting, researchers should consider antibody concentration titration, buffer optimization, and inclusion of appropriate controls to maximize specificity and signal-to-noise ratio.

How can G6PD antibodies be effectively utilized in immunohistochemistry for research applications?

Immunohistochemical detection of G6PD requires careful tissue processing and staining optimization to preserve antigenicity while minimizing background. The following protocol has proven effective for research applications:

Tissue Preparation:

  • Use formalin-fixed, paraffin-embedded (FFPE) tissue sections (4-6 μm thickness)

  • Deparaffinize and rehydrate sections through xylene and graded alcohols

  • Perform heat-induced epitope retrieval using an appropriate basic retrieval solution to expose antigenic sites

Staining Protocol:

  • Block endogenous peroxidase activity with 3% hydrogen peroxide

  • Apply protein block to reduce non-specific binding

  • Incubate with primary anti-G6PD antibody at 1 μg/ml concentration for 1 hour at room temperature

  • Wash thoroughly with buffer to remove unbound antibody

  • Apply appropriate detection system (e.g., HRP-polymer antibody detection)

  • Develop with DAB substrate to produce a brown chromogenic signal

  • Counterstain with hematoxylin for nuclear visualization

  • Dehydrate, clear, and mount slides with permanent mounting medium

Expected Results and Interpretation:

  • Specific G6PD staining should localize primarily to the cytoplasm

  • Liver cancer sections can serve as positive controls due to high G6PD expression

  • Staining intensity may vary based on metabolic activity and tissue type

For advanced research applications, multiplex immunohistochemistry combining G6PD with other markers (such as immune cell markers or metabolic enzymes) can provide valuable insights into the relationship between G6PD expression and the tumor microenvironment.

What are the key considerations for G6PD antibody validation in experimental research?

Rigorous validation of G6PD antibodies is essential for ensuring experimental reproducibility and reliable data interpretation. Key validation considerations include:

Specificity Testing:

  • Western blot analysis to confirm detection of a single band at the expected molecular weight (58-59 kDa for G6PD)

  • Testing across multiple cell lines with known G6PD expression profiles (e.g., A549, MCF-7, Jurkat cells)

  • Validation in G6PD knockdown/knockout models to confirm specificity

  • Peptide competition assays to verify epitope-specific binding

Cross-Reactivity Assessment:

  • Evaluation of potential cross-reactivity with related enzymes in the pentose phosphate pathway

  • Testing across species if the antibody is designed for cross-species applications

  • Inclusion of appropriate negative controls in every experiment

Application-Specific Validation:

  • Optimization for each intended application (Western blot, IHC, flow cytometry)

  • Determination of optimal working concentrations and conditions for each method

  • Assessment of lot-to-lot consistency for reproducible results

Validation ParameterMethodologyExpected OutcomeSignificance
SpecificityWestern blot with controlsSingle band at 58-59 kDaConfirms target recognition
Cell line reactivityTesting in A549, MCF-7 cellsConsistent detectionValidates across cellular contexts
Knockdown verificationsiRNA/shRNA G6PD reductionProportional signal reductionConfirms signal specificity
Tissue reactivityIHC across normal/cancer tissuesCytoplasmic stainingValidates histological applications

Comprehensive validation not only ensures experimental reliability but also facilitates meaningful comparison of results across different studies and laboratories, advancing collective understanding of G6PD biology.

How does G6PD expression differ across cancer types and what methodologies best capture these differences?

Pan-cancer analyses have revealed complex patterns of G6PD expression across tumor types, with important implications for both basic cancer biology and clinical research:

Expression Patterns Across Cancer Types:

  • G6PD is significantly upregulated in most cancer types compared to matched normal tissues

  • The degree of upregulation varies considerably across different cancer types

  • Expression levels correlate with clinical prognosis in multiple cancers

Cancer-Specific Expression Profiles:

  • Liver cancer (HCC): Marked upregulation with correlation to pathological staging

  • Kidney renal clear cell carcinoma (KIRC): Expression positively correlates with disease progression

  • Breast cancer (BRCA): Higher expression associated with advanced pathological stages

  • Kidney renal papillary cell carcinoma (KIRP): Expression increases with disease advancement

Methodological Approaches for Comparative Analysis:

  • Standardized tissue microarrays for simultaneous analysis across multiple cancer types

  • Consistent immunohistochemical protocols with appropriate controls

  • Complementary molecular analyses (qPCR, RNA-seq) to correlate protein with transcript levels

  • Digital pathology with quantitative image analysis for objective expression assessment

What mechanistic roles does G6PD play in cancer cell metabolism and how can these be experimentally investigated?

G6PD functions at a critical metabolic nexus in cancer cells, contributing to multiple aspects of malignant phenotypes through distinct mechanisms:

Metabolic Reprogramming:

  • G6PD serves as the rate-limiting enzyme in the pentose phosphate pathway (PPP)

  • Enhanced PPP activity provides cancer cells with ribose-5-phosphate for nucleotide synthesis

  • This supports the rapid proliferation characteristic of many cancer types

Redox Homeostasis:

  • G6PD generates NADPH, which is crucial for maintaining redox balance

  • NADPH supports glutathione regeneration, protecting cancer cells from oxidative stress

  • This protective mechanism helps cancer cells survive in hostile microenvironments and withstand treatment-induced oxidative damage

Cell Cycle Regulation:

  • Increased G6PD activity is observed during G1 and late S phase

  • This activity correlates with DNA synthesis and replication in cancer cells

  • G6PD upregulation influences cell cycle regulatory proteins

Treatment Resistance Mechanisms:

  • Elevated G6PD activity enhances stress tolerance in cancer cells

  • This contributes to resistance against chemotherapy, radiotherapy, and targeted therapies

  • Specific examples include cisplatin resistance in lung cancer, erlotinib resistance in pancreatic cancer, and doxorubicin resistance in colon cancer

Experimental Approaches for Mechanistic Studies:

  • Genetic manipulation: siRNA/shRNA knockdown or CRISPR-Cas9 editing of G6PD

  • Pharmacological inhibition: Small molecule G6PD inhibitors with appropriate controls

  • Metabolic flux analysis: Isotope tracing to quantify PPP activity

  • Redox state assessment: Measurement of NADPH/NADP+ ratios and ROS levels

  • Combination therapy models: Testing G6PD inhibition alongside standard treatments

These mechanistic insights highlight G6PD as a multifaceted contributor to cancer metabolism and potential vulnerability that can be therapeutically exploited.

How can G6PD antibodies contribute to studying cancer immunotherapy responses?

G6PD antibodies provide valuable tools for investigating the emerging relationship between metabolic pathways and immunotherapy efficacy:

Prognostic Biomarker Applications:

  • G6PD expression levels correlate with immunotherapy outcomes in clinical studies

  • Melanoma and lung cancer patients with low G6PD expression showed better prognosis when treated with immune checkpoint inhibitors (p=0.0473 for melanoma, p=0.0287 for lung cancer)

  • G6PD antibodies enable assessment of expression in patient samples before and during treatment

Tumor Microenvironment Analysis:

  • G6PD expression relates to immune cell infiltration patterns across multiple cancer types

  • Multiplex immunohistochemistry using G6PD antibodies alongside immune cell markers reveals interactions between metabolic features and immune landscape

  • This approach helps characterize metabolic influences on the immunosuppressive tumor microenvironment

Mechanistic Research Applications:

  • G6PD antibodies track expression changes following immunotherapy

  • They enable investigation of how G6PD levels affect immunogenic cell death mechanisms

  • Knockdown or inhibition confirmation in experimental models studying G6PD-immune interactions

Patient PopulationImmunotherapyG6PD StatusClinical OutcomeStatistical Significance
Melanoma (n=42)Immune Checkpoint InhibitorsLow expressionImproved prognosisp=0.0473
Lung cancer (n=30)Immune Checkpoint InhibitorsLow expressionImproved prognosisp=0.0287

These findings suggest that G6PD expression levels may serve as a predictive biomarker for immunotherapy response, potentially guiding treatment decisions and combination strategies. The ability to reliably detect and quantify G6PD using validated antibodies is therefore crucial for translational research in this rapidly evolving field .

How can G6PD antibodies be utilized in studying immunogenic cell death mechanisms?

Recent research has uncovered a fascinating link between G6PD inhibition and the induction of immunogenic cell death (ICD), with G6PD antibodies playing a central role in elucidating these mechanisms:

Monitoring G6PD Inhibition:

  • G6PD antibodies confirm successful knockdown or inhibition in experimental models

  • Western blot and immunofluorescence analyses verify protein reduction following genetic or chemical intervention

  • Quantitative assessment correlates G6PD levels with the degree of intervention

Detecting ICD Marker Dynamics:

  • Following G6PD inhibition, antibody-based assays can detect key ICD markers:

    • High mobility group box 1 (HMGB1) release from dying cells

    • Calreticulin translocation to the plasma membrane

  • Multiplex immunofluorescence combining G6PD and ICD marker antibodies visualizes these events spatially and temporally

Mechanistic Pathway Analysis:

  • G6PD antibodies help elucidate the molecular connections between:

    • G6PD inhibition

    • NADPH reduction

    • Increased oxidative stress

    • Subsequent immunogenic cell death

  • This pathway analysis explains how metabolic perturbation triggers immunological consequences

Translational Research Applications:

  • Immunohistochemistry with G6PD antibodies in patient samples before and after treatment

  • Correlation of G6PD expression with treatment response and immune infiltration

  • Development of predictive models based on G6PD expression patterns

Experimental ApproachG6PD Antibody ApplicationExpected OutcomeKey Controls
G6PD knockdown validationWestern blotReduced band intensityScrambled shRNA control
ICD marker detectionCo-immunofluorescenceCalreticulin membrane localizationKnown ICD inducer control
Tumor section analysisMultiplex IHCG6PD levels with immune infiltrationIsotype control antibodies

These approaches collectively demonstrate how G6PD antibodies serve as essential tools for unraveling the complex interplay between metabolic pathways and immunogenic cell death mechanisms in cancer research .

What methodologies are emerging for targeting G6PD in experimental cancer therapies?

Research into G6PD as a therapeutic target has generated several promising approaches, each requiring specific experimental designs and validation methods:

Genetic Inhibition Strategies:

  • RNA interference approaches (siRNA/shRNA targeting G6PD)

    • Knockdown in melanoma cells demonstrates reduced tumor growth and enhanced immunotherapy sensitivity

    • Verification requires G6PD antibodies for Western blot confirmation

  • CRISPR-Cas9 gene editing for more permanent G6PD knockout

    • Enables long-term studies of G6PD depletion effects

    • Requires thorough validation using G6PD antibodies and enzymatic assays

Chemical Inhibition Approaches:

  • Small molecule G6PD inhibitors with optimized pharmacokinetic properties

    • Selective compounds that target the enzyme's catalytic or regulatory domains

    • Efficacy monitoring through enzymatic assays and G6PD protein level assessment

  • Natural product derivatives with G6PD inhibitory activity

    • Plant-derived compounds being developed as lead structures

    • Structure-activity relationship studies to enhance specificity

Combination Therapy Designs:

  • G6PD inhibition combined with immune checkpoint blockade

    • Significant tumor reduction observed when G6PD-knockdown tumors treated with anti-PD-L1 antibody

    • Remarkable abscopal effect: inhibiting G6PD in only part of tumor lesions increased sensitivity of untreated lesions to immunotherapy

  • G6PD targeting with conventional chemotherapy

    • Potential to overcome resistance mechanisms related to G6PD overexpression

    • Synergistic effects through reducing cancer cells' oxidative stress management capacity

Experimental results demonstrate particularly promising outcomes when G6PD inhibition is combined with immunotherapy. In mouse melanoma models, G6PD knockdown together with anti-PD-L1 treatment produced significant tumor reduction, suggesting this combined approach may overcome resistance mechanisms and enhance therapeutic efficacy .

How can researchers resolve contradictory findings regarding G6PD expression across different experimental models?

Reconciling variable or seemingly contradictory findings about G6PD in cancer research requires systematic methodological approaches:

Standardized Expression Analysis:

  • Implementing validated G6PD antibodies with consistent protocols across studies

  • Complementing antibody-based detection with mRNA analysis and enzymatic activity assays

  • Comprehensive reporting of experimental conditions, antibody details, and detection methods

Context-Dependent Interpretation Frameworks:

  • Acknowledging that G6PD may have different roles depending on:

    • Cancer type and molecular subtype

    • Genetic background and mutational landscape

    • Stage of cancer progression

    • Treatment history and resistance mechanisms

  • Developing multi-parameter models that incorporate these contextual factors

Multi-omics Integration Strategies:

  • Combining proteomics (using G6PD antibodies), transcriptomics, and metabolomics data

  • Integrating G6PD expression with pathway analysis to understand functional consequences

  • Applying systems biology approaches to model context-dependent effects and resolve apparent contradictions

Heterogeneity Analysis Methods:

  • Single-cell analysis techniques to address intratumoral heterogeneity of G6PD expression

  • Spatial transcriptomics or multiplex immunohistochemistry to map G6PD expression patterns

  • Consideration of clonal evolution in expression dynamics over disease progression

Contradiction TypeAnalytical ApproachVerification MethodResolution Strategy
Expression level discrepanciesMulti-method verificationIndependent antibody validationCross-platform data integration
Functional impact variationsContext-specific pathway analysisMetabolic flux measurementIdentification of context-dependent modifiers
Prognostic value differencesStratification by molecular subtypesMultivariate analysisDevelopment of composite biomarker signatures

By implementing these methodological approaches, researchers can better navigate the complexities of G6PD biology across different experimental systems and translate findings more effectively to clinical applications.

What are common pitfalls in G6PD antibody-based assays and how can they be methodically overcome?

Researchers frequently encounter technical challenges when implementing G6PD antibody-based assays. Understanding these pitfalls and their solutions ensures more reliable and reproducible results:

Cross-Reactivity Issues:

  • Pitfall: Antibodies may cross-react with related dehydrogenases or other proteins

  • Solution:

    • Validate specificity using G6PD knockdown/knockout controls

    • Perform peptide competition assays to confirm epitope specificity

    • Include isotype controls in all experiments to identify non-specific binding

Variable Signal Intensity:

  • Pitfall: Inconsistent staining or band intensity across experiments

  • Solution:

    • Standardize protein loading amounts for Western blot applications

    • Maintain antibody aliquots properly to avoid degradation

    • Include validated positive control samples (e.g., A549 or MCF-7 cell lysates) in each experiment

    • Optimize tissue fixation protocols for immunohistochemistry applications

Background Signal Problems:

  • Pitfall: High background obscuring specific G6PD detection

  • Solution:

    • Optimize blocking conditions with systematic testing of agents and durations

    • Titrate primary and secondary antibody concentrations to determine optimal ratios

    • Increase washing duration and frequency between incubation steps

    • For IHC, ensure complete deparaffinization and appropriate antigen retrieval

Antibody Lot Variability:

  • Pitfall: Inconsistent results with different manufacturing lots

  • Solution:

    • Document lot numbers and test new lots against previously validated ones

    • Consider purchasing larger quantities of validated lots when possible

    • Implement standardized quality control procedures for each new lot

Troubleshooting Guide for Common Issues:

IssuePossible CauseSolutionVerification Method
No signal in Western blotInsufficient protein/degraded antibodyIncrease loading/use fresh antibodyInclude known positive control
Multiple bandsCross-reactivity/protein degradationValidate antibody/add protease inhibitorsG6PD knockdown control
Weak IHC stainingInadequate antigen retrievalOptimize retrieval conditionsTest multiple retrieval methods
Non-specific IHC stainingInsufficient blockingIncrease blocking time/titrate antibodyInclude appropriate controls

Implementing these methodical solutions helps researchers overcome technical challenges and obtain more reliable data when working with G6PD antibodies across different experimental platforms.

How can specificity of G6PD antibodies be ensured across different tissue types?

Ensuring G6PD antibody specificity across diverse tissue types requires a systematic validation approach tailored to tissue-specific characteristics:

Comprehensive Validation Strategy:

  • Evaluate antibodies on multi-tissue panels representing various organ systems

  • Include positive control tissues with known high G6PD expression (e.g., liver)

  • Validate performance in both normal and pathological specimens from each tissue type

  • Document tissue-specific optimization parameters for reproducibility

Multi-method Confirmation Approach:

  • Correlate immunohistochemistry results with Western blot analysis of the same tissues

  • Compare antibody-based detection with mRNA expression data from matched samples

  • Verify staining patterns with enzymatic activity assays where feasible

  • This triangulation approach confirms that antibody binding reflects actual G6PD biology

Tissue-Specific Protocol Optimization:

  • Adjust fixation protocols based on tissue composition (e.g., shorter fixation for fatty tissues)

  • Customize antigen retrieval conditions for each tissue type's specific characteristics

  • Determine optimal antibody concentration and incubation times for each tissue context

  • Modify blocking procedures to address tissue-specific sources of background

Biological Control Implementation:

  • When available, use samples from individuals with G6PD deficiency as biological negative controls

  • Include G6PD knockdown tissues from appropriate experimental models

  • Consider tissue-specific conditional knockout models for definitive validation

Tissue TypeExpected G6PD ExpressionOptimization RequirementsPotential Interfering Factors
Liver tissueHigh (metabolically active)Mild antigen retrievalEndogenous peroxidases, glycogen
Cancer specimensVariable, often elevatedCase-by-case adjustmentNecrotic areas, heterogeneity
Blood cellsVariable (affected in G6PD deficiency)Specialized fixation methodsHemoglobin interference
Brain tissueModerate expressionExtended fixation, specific retrievalLipid content affects antibody penetration

What are the recommended controls for G6PD antibody experiments in different research applications?

Implementing appropriate controls is essential for ensuring reliable and interpretable results with G6PD antibodies. The following controls should be considered for different experimental applications:

Positive Controls:

  • Cell lines with validated G6PD expression (e.g., A549, MCF-7, Jurkat cells as demonstrated in research)

  • Tissue sections known to express G6PD at high levels (e.g., liver tissue)

  • Recombinant G6PD protein standards for quantitative applications

  • These controls confirm that detection systems are functioning properly

Negative Controls:

  • G6PD knockdown or knockout cell lines/tissues when available

  • Samples from individuals with severe G6PD deficiency (for clinical research)

  • Primary antibody omission controls to assess non-specific binding of detection systems

  • Isotype controls using irrelevant antibodies of the same isotype and concentration

Specificity Controls:

  • Peptide competition/blocking with the immunizing peptide

  • Testing multiple antibodies targeting different G6PD epitopes

  • Correlation with G6PD mRNA expression or enzymatic activity measurements

  • These controls verify that the signal detected truly represents G6PD protein

Technical Controls for Different Applications:

ApplicationEssential ControlsPurposeImplementation
Western BlotLoading control, molecular weight markerEnsure equal loading, confirm target identityInclude on every blot
IHCPositive tissue, negative control tissueVerify protocol functionality, identify non-specific bindingProcess in same batch
Flow CytometryUnstained cells, isotype controlSet gates properly, control for backgroundInclude in each run
Simple Western™Concentration gradient, positive controlEstablish quantitative range, verify system functionRun on same array

Experimental Design Controls:

  • Biological replicates (samples from different sources)

  • Technical replicates (multiple analyses of the same sample)

  • Time course controls to assess stability of G6PD expression during experimental manipulation

By systematically implementing these controls, researchers can substantiate their findings, troubleshoot technical issues effectively, and produce more reliable and reproducible data when working with G6PD antibodies across different experimental systems.

Product Science Overview

Introduction

Glucose-6-Phosphate Dehydrogenase (G6PD) is a crucial enzyme in the pentose phosphate pathway, which is essential for cellular metabolism. This enzyme catalyzes the conversion of glucose-6-phosphate into 6-phosphoglucono-δ-lactone while reducing NADP+ to NADPH. The NADPH produced is vital for protecting cells against oxidative damage by maintaining the level of reduced glutathione .

Structure and Function

G6PD is a cytosolic enzyme that plays a significant role in the pentose phosphate pathway, which provides reducing energy to cells by maintaining the level of NADPH. NADPH, in turn, helps in maintaining the level of reduced glutathione, which protects red blood cells from oxidative damage . The enzyme is also involved in the Entner-Doudoroff pathway, a type of glycolysis .

Clinical Significance

G6PD deficiency is an X-linked genetic disorder that affects millions of people worldwide. This deficiency makes individuals prone to non-immune hemolytic anemia, especially under oxidative stress conditions such as infections, certain medications, or ingestion of fava beans . The deficiency is particularly prevalent in regions where malaria is or was common, as the condition provides some protection against malaria .

Mouse Anti Human G6PD

Mouse anti-human G6PD antibodies are used in various research and diagnostic applications. These antibodies are typically produced by immunizing mice with human G6PD protein, leading to the generation of monoclonal or polyclonal antibodies that specifically recognize human G6PD . These antibodies can be used in techniques such as Western blotting, immunohistochemistry, and enzyme-linked immunosorbent assays (ELISA) to detect and quantify G6PD in human samples .

Research Applications

Humanized mouse models of G6PD deficiency have been developed to study the effects of this condition in vivo. These models involve the introduction of human G6PD genes into mice, allowing researchers to study the physiological and biochemical impacts of G6PD deficiency under controlled conditions . Such models are invaluable for testing the hemolytic potential of drugs and understanding the molecular mechanisms underlying G6PD deficiency .

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.