Produced in Escherichia coli as a single, non-glycosylated polypeptide chain containing 356 amino acids (residues 1-333 + 23-amino acid His-tag) .
Purification: Proprietary chromatographic techniques ensure >95% purity .
GAPDH Mouse, Active demonstrates dual enzymatic activities:
Glycolytic role: Catalyzes the reversible conversion of glyceraldehyde-3-phosphate to 1,3-bisphosphoglycerate using NAD⁺ .
Nitrosylase activity: Mediates S-nitrosylation of nuclear proteins like SIRT1 and HDAC2, influencing epigenetic regulation .
Additional non-glycolytic functions include:
Cytoskeleton organization via CHP1-microtubule interactions .
Immune modulation through TRAF2/3 binding to enhance NF-κB activation and type I interferon production .
G93A and H46R/H48Q mouse models: GAPDH shows 41-43% reduced enzymatic activity due to oxidative stress-induced conformational changes. Surface hydrophobic domains are altered, impairing function without affecting protein levels .
Parameter | G93A Mice vs. NTg | H46R/H48Q Mice vs. NTg |
---|---|---|
GAPDH activity | ↓43% | ↓41% |
Conformational changes | Novel hydrophobic domains observed | Similar alterations |
Pre-treatment with 10 mg/kg GAPDH increased survival rates from 0% to 80% at 48 hours post-LPS challenge .
Mechanisms include:
Outcome Metric | GAPDH Pre-Treatment | PBS Control |
---|---|---|
Survival rate (48h) | 80% | 0% |
Serum IL-6 (12h) | 120 pg/mL | 400 pg/mL |
Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a key enzyme involved in glycolysis, catalyzing the reversible oxidative phosphorylation of glyceraldehyde-3-phosphate in the presence of inorganic phosphate and nicotinamide adenine dinucleotide (NAD). The enzyme exists as a tetramer of identical chains and plays a critical role in cellular energy metabolism . Beyond its glycolytic function, GAPDH has been identified as a "moonlighting protein" with numerous non-glycolytic roles including membrane fusion, microtubule bundling, phosphotransferase activity, nuclear RNA export, DNA replication, and DNA repair . GAPDH also contains a peptide with antimicrobial activity against several microorganisms including E. coli, P. aeruginosa, and C. albicans . Additionally, in mouse models, GAPDH has been implicated in the regulation of mRNA stability, nitrosylation of nuclear proteins, and can act as a transferrin receptor on macrophage cell surfaces .
Mouse GAPDH has a calculated molecular weight of 36 kDa, though it typically appears as a band at approximately 38-39 kDa in Western blot applications due to post-translational modifications . GAPDH can be detected using various complementary techniques. In Western blotting, specific antibodies such as Mouse Anti-Human/Mouse/Rat GAPDH/G3PDH Monoclonal Antibody detect a specific band at approximately 39 kDa in mouse brain tissue lysates . For immunohistochemistry (IHC), GAPDH can be visualized in paraffin-embedded tissue sections using appropriate antibodies, often requiring heat-induced epitope retrieval for optimal results . In cellular immunofluorescence studies, GAPDH typically shows both cytoplasmic and nuclear localization patterns . For gene expression analysis, readymade GAPDH primers are available that work effectively for mouse GAPDH, generating an amplicon size of 452 bp .
When using mouse GAPDH as a loading control in Western blots, researchers should implement several best practices to ensure reliable results. First, select antibodies specifically validated for mouse GAPDH, such as monoclonal antibodies that detect a band at approximately 39 kDa or polyclonal antibodies that detect a band at approximately 38 kDa . Determine the optimal antibody concentration for your specific application; some anti-GAPDH antibodies may be used at dilutions as high as 1:100,000 to 1:660,000 for Western blots due to GAPDH's abundant expression . Due to this high abundance, using appropriate amounts of total protein (typically 10-20 μg) is crucial to avoid signal saturation which can compromise quantitative analysis. Always validate that the GAPDH signal falls within the linear range of detection by running a standard curve with different amounts of the same sample. Consider that certain experimental treatments might affect GAPDH expression; in such cases, alternative loading controls or total protein staining methods might be more appropriate. Finally, maintain consistent experimental conditions including reducing agents and buffer systems; for example, Immunoblot Buffer Group 2 has been reported to yield good results for GAPDH detection .
For reliable detection of GAPDH in mouse samples, researchers should select validated primer sets that offer high specificity and efficiency. ReadyMade GAPDH primers are commercially available that work effectively in mouse samples, generating an amplicon size of 452 bp . These primers are designed to work with both human and mouse GAPDH, though they have a single base mismatch with rat GAPDH . When selecting primers, consider the application requirements—for standard PCR, longer amplicons (400-500 bp) may be acceptable, while qPCR typically benefits from shorter amplicons (70-150 bp) for optimal efficiency. For splice variant analysis, design primers that span exon-exon junctions to avoid genomic DNA amplification and to distinguish between specific variants. Always validate new primer sets by checking amplification efficiency (ideally between 90-110%), specificity (single peak in melt curve analysis), and linear range of detection across relevant expression levels. For absolute quantification, generate standard curves using plasmids containing the mouse GAPDH sequence. When comparing GAPDH across multiple tissues or experimental conditions, verify primer performance in each specific context, as primer efficiency can vary with sample composition.
Mouse GAPDH undergoes various post-translational modifications that significantly alter its enzymatic activity, subcellular localization, and protein-protein interactions. S-nitrosylation of cysteine residues, particularly Cys-150, can inhibit GAPDH's glycolytic activity while promoting its binding to Siah1 (an E3 ubiquitin ligase), leading to nuclear translocation. This modification mechanism has been implicated in cell death pathways in mouse models of neurodegenerative diseases . Phosphorylation by various kinases including AMP-activated protein kinase (AMPK) affects both GAPDH's enzymatic activity and its non-glycolytic functions, particularly in autophagy regulation. Acetylation of lysine residues modulates GAPDH's interaction with other proteins and influences its nuclear translocation, while O-GlcNAcylation occurs in response to changes in glucose availability and can alter both GAPDH's enzymatic activity and subcellular distribution. These modifications provide molecular mechanisms through which cells can regulate the diverse functions of GAPDH in response to various stimuli and metabolic states. When designing experiments to study GAPDH activity, researchers should consider how their experimental conditions might affect these modifications and include assays that can detect specific post-translational states.
GAPDH has been identified as a multifunctional "moonlighting" protein in mouse models, exhibiting numerous non-glycolytic functions beyond its classical role in energy metabolism . In the nucleus, GAPDH exhibits uracil DNA glycosylase activity, participating in DNA repair mechanisms, while also playing roles in the nitrosylation of nuclear proteins and regulation of mRNA stability . At the cell surface, GAPDH can act as a transferrin receptor on macrophages, potentially contributing to iron homeostasis . Mouse GAPDH contains peptides with demonstrated antimicrobial activity against several microorganisms including E. coli, P. aeruginosa, and C. albicans, suggesting a role in innate immunity . Within the cytoplasm, GAPDH participates in membrane fusion processes and microtubule bundling, contributing to cellular structure and trafficking regulation . Nuclear translocation of GAPDH has been linked to programmed cell death pathways, particularly in neuronal cells, making it relevant for neurodegenerative disease research . Recent research also suggests that GAPDH can enhance tight junction regeneration after damage, indicating a potential role in cellular barrier function and repair processes .
GAPDH has been implicated in various neurodegenerative disease processes in mouse models, particularly through mechanisms involving its non-glycolytic functions. In models of Alzheimer's disease, GAPDH has been found to interact with amyloid-β and tau proteins, with nuclear accumulation of GAPDH correlating with neuronal loss . Studies have shown that preventing this nuclear translocation can have neuroprotective effects in these models. For Huntington's disease, GAPDH can bind to mutant huntingtin protein, potentially exacerbating protein aggregation and cytotoxicity. Mouse models of Huntington's disease consistently show altered GAPDH function and abnormal subcellular localization . In MPTP-induced mouse models of Parkinson's disease, GAPDH has been linked to dopaminergic neuronal death through nitrosylation-dependent mechanisms. More broadly, GAPDH plays a key role in the neuronal response to oxidative damage, a common factor in various neurodegenerative conditions. Its nuclear translocation under oxidative stress conditions can trigger apoptotic pathways that contribute to neurodegeneration. These findings highlight the importance of considering GAPDH not just as a housekeeping gene but as an active participant in pathological processes when using mouse models to study neurodegenerative diseases .
Differentiating between the glycolytic and non-glycolytic functions of mouse GAPDH requires carefully designed experimental approaches. Site-directed mutagenesis can generate mouse models or cell lines expressing GAPDH variants with mutations in specific functional domains. For example, mutations in the catalytic site (around Cys-150) can disrupt glycolytic activity while potentially preserving other functions. Subcellular localization analysis is particularly valuable since glycolytic GAPDH functions primarily in the cytoplasm, while many non-glycolytic functions occur in the nucleus or at the cell surface. Tracking localization through fractionation and immunostaining can help distinguish these roles . In some experimental systems, glycolytic and non-glycolytic functions can be temporally separated—rapid responses to glucose fluctuations are likely related to glycolytic roles, while delayed responses might involve gene regulation or other non-metabolic functions. Selective inhibitors targeting either GAPDH's enzymatic activity or its interactions with particular binding partners can help isolate specific functions. Correlation analysis between enzymatic activity (measured through standard dehydrogenase assays) and other cellular outcomes can determine whether effects are likely mediated by glycolytic or non-glycolytic mechanisms. Finally, interactome analysis identifying proteins associating with GAPDH under different conditions can provide insights into which functional pathways are being engaged.
For accurate measurement of mouse GAPDH enzymatic activity, researchers should optimize several key parameters. The optimal buffer composition typically includes pH 8.5-8.6 phosphate or Tris buffer, 1-2 mM NAD+ as a cofactor, 5-10 mM sodium arsenate or inorganic phosphate, and 1-2 mM DTT or β-mercaptoethanol to maintain reducing conditions. Temperature control is crucial, with 25-37°C being the typical range and 30°C often providing an ideal balance between enzyme stability and reaction rate. Substrate concentration should be carefully optimized, with 0.5-2 mM glyceraldehyde-3-phosphate typically ensuring the reaction remains in the linear range for accurate measurements. Sample preparation requires tissues to be homogenized in cold buffer containing protease inhibitors to prevent degradation, with brief sonication potentially improving enzyme extraction efficiency. For measurement approaches, researchers can employ spectrophotometric assays monitoring the increase in absorbance at 340 nm due to NADH formation, or use fluorometric assays measuring NADH fluorescence (excitation 340 nm, emission 460 nm) for increased sensitivity. Controls and calibration standards are essential, including known concentrations of purified GAPDH as positive controls, enzyme kinetics analysis (Km and Vmax determination), and samples with GAPDH inhibitors as negative controls to verify assay specificity.
Despite its common use as a reference gene, GAPDH expression can vary significantly under certain experimental conditions. To account for these variations when using mouse GAPDH as a reference gene in qPCR, researchers should first validate its stability under their specific experimental conditions by comparing expression across all treatment groups and time points. Following MIQE guidelines, it's advisable to use GAPDH in combination with other reference genes (such as β-actin, 18S rRNA, or HPRT) and calculate a normalization factor based on their geometric mean rather than relying on a single reference gene. Statistical algorithms such as geNorm, NormFinder, or BestKeeper can help determine the most stable reference genes for specific experimental conditions. Using validated GAPDH primers that work efficiently in mouse samples is essential; readymade primers are available that generate a 452 bp amplicon in mouse samples . Always determine the amplification efficiency of GAPDH primers and incorporate this into calculations rather than assuming 100% efficiency. Be especially aware that GAPDH expression varies significantly between different mouse tissues and can be directly affected by experimental treatments that influence glycolysis or related metabolic pathways. Through careful validation and appropriate normalization strategies, researchers can minimize the impact of GAPDH variations on their qPCR results.
Based on the available data, several antibodies have demonstrated high effectiveness for detecting mouse GAPDH across different applications. For Western blotting, Mouse Anti-Human/Mouse/Rat GAPDH/G3PDH Monoclonal Antibody (Clone #686613) works effectively at very low concentrations (0.05 μg/mL) and detects a specific band at approximately 39 kDa in mouse brain tissue . Alternatively, Goat Anti-Human/Mouse/Rat GAPDH/G3PDH Antigen Affinity-purified Polyclonal Antibody used at 1 μg/mL detects a specific band at approximately 38 kDa . For high-sensitivity applications, Mouse mAb (High Dilution) can be used at extremely high dilutions (1:100,000 - 1:660,000) due to its exceptional titer . For immunohistochemistry on paraffin-embedded sections (IHC-P), Mouse Anti-Human/Mouse/Rat GAPDH/G3PDH Monoclonal Antibody has shown good results at 15 μg/mL when combined with heat-induced epitope retrieval , while Goat Anti-Human/Mouse/Rat GAPDH/G3PDH Antigen Affinity-purified Polyclonal Antibody works well at 8 μg/mL with overnight incubation at 4°C . For immunofluorescence applications, both monoclonal (8 μg/mL) and polyclonal (5 μg/mL) antibodies have demonstrated effective staining of GAPDH in fixed cells with 3-hour room temperature incubations . When selecting antibodies, researchers should consider the specific application requirements, whether monoclonal specificity or polyclonal sensitivity is more important, required species cross-reactivity, and validation status in the specific mouse strain or tissue of interest.
To effectively study GAPDH's roles in specific cellular compartments in mouse models, several complementary experimental approaches are recommended. Subcellular fractionation combined with Western blotting allows separation of nuclear, cytoplasmic, mitochondrial, and membrane fractions using differential centrifugation, followed by GAPDH detection with specific antibodies . This approach should include compartment-specific markers to verify fractionation purity. Immunofluorescence microscopy with co-localization analysis provides spatial information by co-staining cells or tissue sections with anti-GAPDH antibodies and markers for specific organelles. Confocal or super-resolution microscopy enables precise localization, with quantitative co-localization analysis using Pearson's or Mander's coefficients. As shown in previous studies, GAPDH can be successfully visualized in both cytoplasm and nuclei using this approach . For dynamic studies, proximity labeling in living cells can be achieved by generating mouse cell lines expressing GAPDH fused to promiscuous biotin ligases or peroxidases, allowing identification of proteins near GAPDH in specific compartments. Compartment-targeted GAPDH variants with additional targeting sequences can help assess compartment-specific functions. Live-cell techniques such as Fluorescence Recovery After Photobleaching (FRAP) with fluorescently-tagged GAPDH provide insights into dynamic movements between compartments under different cellular conditions. When implementing these designs, researchers should carefully consider how sample preparation might affect GAPDH localization and ensure that any tags do not interfere with native functions.
Modulating GAPDH expression in mouse models requires careful consideration of its essential metabolic functions and potential compensatory mechanisms. For knockdown approaches, conditional/inducible systems are strongly recommended, as complete GAPDH knockout is often lethal. Doxycycline-inducible shRNA systems or Cre-loxP configurations enable temporal and tissue-specific control of GAPDH reduction. Partial knockdown strategies using siRNA or shRNA constructs achieving moderate (50-70%) reduction in GAPDH levels can maintain cellular viability while revealing phenotypic effects. Validation of knockdown efficiency should be performed by Western blot using specific antibodies as described in previous studies . For overexpression strategies, viral vector systems such as adeno-associated virus (AAV) with tissue-specific promoters or lentiviral vectors for stable integration provide efficient delivery methods. When designing overexpression constructs, consider including epitope tags or fluorescent proteins for easy detection, but verify these tags don't interfere with GAPDH function. For transient or therapeutic applications, formulated modified GAPDH mRNA in lipid nanoparticles offers controlled temporary expression with reduced genomic integration risk. For all approaches, comprehensive validation should include both mRNA and protein analysis, assessment of effects on glycolytic function and cell viability, appropriate controls, and monitoring of potential compensatory changes in related genes or off-target effects.
Contradictory data regarding GAPDH expression across mouse tissues can arise from both technical and biological variables. To resolve such discrepancies, researchers should implement several systematic approaches. Standardization of tissue collection and processing is paramount—harvest tissues at consistent times of day to account for circadian variations, control the nutritional status of mice, and process all comparative samples simultaneously to minimize batch effects. Employing multiple detection methods provides more robust evidence; compare results from qPCR, Western blotting using multiple validated antibodies targeting different GAPDH epitopes , and immunohistochemistry. Comprehensive statistical analysis with increased biological replicates improves statistical power and helps identify potential outliers or technical artifacts. The assumption that GAPDH is stably expressed across all tissues should be challenged by validating reference genes for each specific tissue type using algorithms like NormFinder or geNorm. Tissue heterogeneity must be considered, as cell type-specific expression patterns might be masked in whole-tissue analyses; techniques like laser capture microdissection or single-cell approaches can reveal these differences. Potential isoform-specific expression should be evaluated by designing assays that can distinguish between GAPDH variants. Finally, a meta-analysis approach systematically reviewing published literature can identify methodological factors explaining contradictory results and lead to standardized protocols based on consensus findings.
Interpreting GAPDH Western blot results in mouse studies involves navigating several common pitfalls that can compromise data quality. Signal saturation frequently occurs due to GAPDH's high abundance, flattening band intensity differences and compromising quantitative comparisons. This can be addressed by using lower protein amounts, highly diluted primary antibodies (some can be used at dilutions as high as 1:660,000) , or shorter exposure times. Inconsistent loading is another major issue when using GAPDH as a loading control; verify equal loading with total protein stains before GAPDH detection to ensure observed variations represent true biological differences. GAPDH expression naturally varies across mouse tissues, necessitating validation as an appropriate control for specific tissues and experimental conditions. Post-translational modifications can alter antibody binding or cause band shifts; using antibodies that detect GAPDH regardless of modification state or employing multiple antibodies targeting different epitopes helps address this issue . Antibody cross-reactivity with other dehydrogenases or similarly sized proteins can be verified using appropriate controls. Protein degradation artifacts creating additional bands can be minimized by maintaining strict sample handling protocols and verifying a single band of appropriate molecular weight (approximately 38-39 kDa for mouse GAPDH) . Membrane and transfer inconsistencies causing regional variations in signal can be controlled using internal lane markers and verification of transfer efficiency. Quantification challenges related to background subtraction methods can significantly impact results and should be addressed through consistent protocols.
Proper normalization of GAPDH data across different experimental conditions is essential for accurate interpretation and comparison. When GAPDH is the target protein being studied, total protein normalization provides a robust approach—stain membranes with total protein stains and normalize GAPDH signal to total protein in each lane. This accounts for loading differences without relying on a single reference protein that might vary across conditions. For higher reliability, implement the multiple reference protein approach using several stable reference proteins and calculating a normalization factor based on their geometric mean. This minimizes the impact of variation in any single reference protein. For absolute quantification, include a standard curve of purified GAPDH protein to calculate absolute amounts rather than relative values, enabling direct comparison across different experiments. When using GAPDH as a reference gene in qPCR, first validate its stability using algorithms like geNorm or NormFinder to assess suitability for your conditions. Follow MIQE guidelines by combining GAPDH with other reference genes and using their geometric mean for normalization. Always determine PCR efficiency for GAPDH amplification in your specific samples and apply efficiency correction in quantification formulas. For immunohistochemistry or immunofluorescence studies, normalize GAPDH staining to unchanging anatomical features and consider analyzing nuclear-to-cytoplasmic GAPDH ratios, which can reveal redistribution effects even when total levels remain similar. This compartment analysis is particularly relevant since GAPDH has been observed in both cytoplasmic and nuclear locations across multiple studies .
Implementing rigorous quality control measures when using GAPDH antibodies is essential for generating reliable and reproducible data in mouse studies. Begin with antibody validation through multiple detection methods—verify that the antibody detects a single band of the expected molecular weight (approximately 38-39 kDa) in Western blots of mouse tissues , shows expected localization patterns in immunostaining, and demonstrates appropriate species cross-reactivity. Include appropriate positive controls (tissues or cell types known to express GAPDH) and negative controls (secondary antibody-only staining or samples with GAPDH knockdown) in every experiment. Perform titration experiments to determine the optimal antibody concentration for each specific application; antibody requirements can vary dramatically, with some GAPDH antibodies effective at dilutions as high as 1:660,000 for Western blotting versus 1:50-1:200 for immunofluorescence. Verify antibody specificity through peptide competition assays where pre-incubation with the immunizing peptide should abolish specific staining. For quantitative applications, confirm that detection falls within the linear range by creating standard curves with serial dilutions of sample. Batch-to-batch variation should be monitored by including reference samples across different antibody lots. For application-specific considerations, verify that antibodies used for immunohistochemistry are compatible with your fixation method and that those for flow cytometry recognize native epitopes. Finally, comprehensive documentation of antibody details (manufacturer, catalog number, lot number, dilution, incubation conditions) ensures experimental reproducibility and facilitates troubleshooting of unexpected results.
Many experimental treatments directly affect GAPDH expression or activity, potentially confounding its use as a normalization control. To mitigate these effects, researchers should first conduct preliminary validation experiments assessing GAPDH stability under specific experimental conditions before relying on it as a reference. Implementing a multiple reference gene approach significantly improves reliability—combine GAPDH with other housekeeping genes and validate their collective stability using algorithms like geNorm or NormFinder. For protein-level normalization, consider alternatives to single protein loading controls such as total protein normalization methods (Ponceau S, SYPRO Ruby, or stain-free technologies), which are less affected by treatments that alter specific proteins. When experimental conditions are known to affect GAPDH expression, implement absolute quantification approaches using standard curves of recombinant GAPDH protein rather than relative quantification methods. For treatments that alter GAPDH post-translational modifications, select antibodies recognizing epitopes unaffected by these modifications or use multiple antibodies targeting different regions of the protein . Design experimental protocols with appropriate randomization and include untreated control samples processed alongside treated samples in every analytical batch. For longitudinal studies or those comparing multiple treatment conditions, include cross-condition calibration samples to allow for batch correction during data analysis. By implementing these strategies, researchers can generate more reliable comparative data even when working with experimental conditions that potentially regulate GAPDH expression or function.
GAPDH is a tetrameric enzyme composed of four identical subunits, each with a molecular weight of approximately 36 kDa . The recombinant mouse GAPDH protein is typically expressed in Escherichia coli and purified to a high degree of purity (>95%) using conventional chromatography techniques . The protein sequence includes a His tag at the N-terminus to facilitate purification .
GAPDH plays a dual role in cellular metabolism:
The specific activity of the recombinant mouse GAPDH is greater than 40 units/mg, defined as the amount of enzyme that converts 1.0 µmole of glyceraldehyde-3-phosphate to 1,3-bisphosphoglycerate per minute at pH 8.5 at 37°C .
Due to its critical role in both glycolysis and nuclear functions, GAPDH is a valuable tool in various research applications: