Recombinant Mouse Alanine aminotransferase 2 (Gpt2)

Shipped with Ice Packs
In Stock

Description

Biological Function and Metabolic Roles

GPT2 operates in mitochondrial metabolism, bridging amino acid and carbohydrate pathways:

Core Enzymatic Activity

  • Reaction:

    L-Alanine+2-OxoglutarateL-Glutamate+Pyruvate\text{L-Alanine} + \text{2-Oxoglutarate} \rightleftharpoons \text{L-Glutamate} + \text{Pyruvate}
  • Catalytic Efficiency: Pyridoxal-phosphate-dependent, with high substrate specificity .

Metabolic Pathways

PathwayRoleReferences
Alanine MetabolismMaintains alanine homeostasis; regulates glutamate synthesis.
TCA Cycle AnaplerosisSupplies α-ketoglutarate for TCA cycle intermediates.
Glutamine UtilizationFacilitates glutamine-to-glutamate conversion in hypoxic conditions.

Neurological Disorders

  • GPT2 Deficiency: Loss-of-function mutations cause intellectual disability, microcephaly, and spastic paraplegia. Gpt2-null mice exhibit reduced brain growth and abnormal metabolite profiles (e.g., low alanine, elevated phenylalanine) .

  • Mechanistic Insights: Defective GPT2 disrupts amino acid metabolism and TCA cycle anaplerosis, impairing neuroprotective pathways .

Cancer Biology

  • Hypoxia-Induced Expression: GPT2 is upregulated in glioblastoma (GBM) via HIF-2α, promoting cell migration and tumor growth. Inhibition reduces colony formation in GBM models .

  • Metabolic Adaptation: Supports glutamine-dependent anaplerosis in hypoxic tumors, enhancing survival .

Experimental Uses

ApplicationDescriptionReferences
Enzyme Activity AssaysQuantify transamination kinetics in vitro.
Disease ModelingStudy GPT2-linked encephalopathy and cancer progression in murine models.
Antibody DevelopmentValidate GPT2 expression in tissues (e.g., IHC, Western blot) .

Therapeutic Relevance

  • Target for Metabolic Disorders: Modulating GPT2 may address neurological deficits or cancer metabolism .

  • Biomarker Potential: Elevated GPT2 in GBM tissues suggests diagnostic utility .

Production and Quality Control

ParameterSpecificationReferences
Storage-20°C for long-term; avoid freeze-thaw cycles. Add BSA/HSA for stability.
Contaminant ControlEndotoxin levels <1 EU/µg (Abcam) .
Functional ValidationSDS-PAGE, mass spectrometry, and activity assays confirm integrity.

Product Specs

Form
Lyophilized powder Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized fulfillment.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates. Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notification and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to pellet the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing. The tag type is determined during production. If a specific tag type is required, please inform us for preferential development.
Synonyms
Gpt2; Aat2Alanine aminotransferase 2; ALT2; EC 2.6.1.2; Glutamate pyruvate transaminase 2; GPT 2; Glutamic--alanine transaminase 2; Glutamic--pyruvic transaminase 2
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-522
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Mus musculus (Mouse)
Target Names
Gpt2
Target Protein Sequence
MQRAAVLVRR GSCPRASGPW GRSHSSAAAE ASAALKVRPE RSPRDRILTL ESMNPQVKAV EYAVRGPIVL KAGEIEMELQ RGIKKPFTEV IRANIGDAHA MGQQPITFLR QVMALCTYPN LLNSPSFPED AKKRARRILQ ACGGNSLGSY SASQGVNCIR EDVAAFITRR DGVPADPDNI YLTTGASDGI STILKLLVSG GGKSRTGVMI PIPQYPLYSA VISELDAVQV NYYLDEENCW ALNVDELRRA LRQAKDHCDP KVLCIINPGN PTGQVQSRKC IEDVIHFAWE EKLFLLADEV YQDNVYSPDC RFHSFKKVLY QMGHEYSSNV ELASFHSTSK GYMGECGYRG GYMEVINLHP EIKGQLVKLL SVRLCPPVSG QAAMDIVVNP PEPGEESFEQ FSREKEFVLG NLAKKAKLTE DLFNQVPGIQ CNPLQGAMYA FPRILIPAKA VEAAQSHKMA PDMFYCMKLL EETGICVVPG SGFGQREGTY HFRMTILPPV DKLKTVLHKV KDFHLKFLEQ YS
Uniprot No.

Target Background

Function

Catalyzes the reversible transamination between alanine and 2-oxoglutarate, yielding pyruvate and glutamate.

Gene References Into Functions
  1. Increased GPT2 gene expression in the brain during early postnatal development, with mitochondrial localization of the GPT2 protein. Similar to the human phenotype, Gpt2-null mice exhibit reduced brain growth. Metabolomic and isotope tracing studies reveal several metabolic abnormalities linked to Gpt2 deficiency. PMID: 27601654
  2. Alt2/Gpt2 (but not Alt1/Gpt1) shows induced expression in the liver of obese male ob/ob mice compared to lean C57BL/6J mice. PMID: 25865565
Database Links
Protein Families
Class-I pyridoxal-phosphate-dependent aminotransferase family, Alanine aminotransferase subfamily
Tissue Specificity
Specifically induced in fatty liver. Highly expressed in muscle, liver and white adipose tissue. Moderately expressed in brain and kidney and expressed at low levels in the heart.

Q&A

What is mouse GPT2 and what are its primary functions in cellular metabolism?

Mouse Glutamic-Pyruvic Transaminase 2 (GPT2) is a mitochondrial alanine transaminase that catalyzes the reversible transamination between alanine and 2-oxoglutarate to form pyruvate and glutamate. This enzyme plays crucial roles in gluconeogenesis and amino acid metabolism, particularly in skeletal muscle, kidney, and adipose tissue. Unlike its paralog GPT1 (which is predominantly cytosolic and liver-expressed), GPT2 is primarily expressed in muscle, fat, and kidney tissues, where it participates in the intermediary metabolism of glucose and amino acids . The enzyme functions as a pyridoxal phosphate-dependent enzyme and is integral to maintaining nitrogen balance and energy homeostasis across multiple tissue types. Its activity becomes particularly important during metabolic stress conditions, when activating transcription factor 4 has been shown to upregulate this gene in hepatocyte cell lines .

What is the molecular structure of recombinant mouse GPT2 protein?

Recombinant mouse GPT2 protein produced in E. coli is a single, non-glycosylated polypeptide chain containing 522 amino acids (positions 1-522) with a molecular mass of approximately 60.1 kDa. For research applications, it is typically fused to a 20-21 amino acid His-tag at the N-terminus to facilitate purification using affinity chromatography techniques . The native protein functions as a homodimer and contains binding sites for its substrate alanine, the coenzyme pyridoxal phosphate, and 2-oxoglutarate. The protein's 3D structure features characteristic domains of aminotransferases, including catalytic and cofactor-binding regions that are essential for its enzymatic activity. The recombinant version maintains the biological activity of native GPT2, with specific activity exceeding 50 units/mg when measured by the amount of enzyme that cleaves 1 μmole of L-Alanine to L-Glutamate per minute at pH 7.5 at 37°C .

How does recombinant mouse GPT2 differ from mouse GPT1?

While GPT1 and GPT2 catalyze the same biochemical reaction, they differ significantly in their cellular localization, tissue distribution, and physiological roles. GPT1 is predominantly cytosolic and highly expressed in liver tissue, while GPT2 is mitochondrial and more broadly expressed in muscle, adipose tissue, and kidneys . Recombinant mouse GPT2 has a higher molecular weight (60.1 kDa) compared to GPT1. From a functional perspective, GPT2 is more associated with amino acid metabolism in non-hepatic tissues and responds differently to metabolic stressors. When designing experiments, researchers should consider these differences, as they influence the interpretation of data, especially in metabolic studies. The amino acid sequence homology between GPT1 and GPT2 is approximately 65-70%, with key differences in the mitochondrial targeting sequence present only in GPT2. These structural differences result in distinct kinetic properties, with GPT2 typically showing different substrate affinities than GPT1, information that is critical for experimental design when studying isoform-specific functions .

What is the optimal storage protocol for recombinant mouse GPT2?

For maximum stability and retention of enzymatic activity, recombinant mouse GPT2 should be stored according to the following protocol: For short-term storage (2-4 weeks), the protein can be kept at 4°C. For long-term storage, the protein should be stored at -20°C . To prevent protein degradation over extended periods, it is strongly recommended to add a carrier protein such as 0.1% Human Serum Albumin (HSA) or Bovine Serum Albumin (BSA) . Multiple freeze-thaw cycles should be strictly avoided as they significantly reduce enzymatic activity through protein denaturation. If frequent use is anticipated, prepare small aliquots of the protein solution before freezing to minimize freeze-thaw cycles. The standard formulation of recombinant mouse GPT2 (1 mg/ml) contains 20 mM Tris-HCl buffer (pH 7.5), 20% glycerol as a cryoprotectant, and 2 mM DTT to maintain the protein's reduced state . Any deviation from these storage conditions should be carefully validated before implementing in research protocols.

What purification methods are most effective for recombinant mouse GPT2?

Recombinant mouse GPT2 is typically purified using a multi-step chromatographic approach, capitalizing on the engineered His-tag at its N-terminus. The most effective purification protocol involves:

  • Initial capture: Immobilized Metal Affinity Chromatography (IMAC) using Ni-NTA or similar matrices to bind the His-tagged protein with high specificity.

  • Intermediate purification: Ion exchange chromatography to separate the target protein from remaining contaminants based on charge differences.

  • Polishing step: Size exclusion chromatography to achieve final purity by separating monomeric GPT2 from aggregates and remaining impurities.

For research requiring higher purity than the standard >85% (as determined by SDS-PAGE) , additional purification steps may be necessary. The purified protein should be maintained in a stabilizing buffer containing 20 mM Tris-HCl (pH 7.5), 20% glycerol, and 2 mM DTT . When designing a purification strategy, researchers should consider that GPT2 activity requires pyridoxal phosphate as a cofactor, which may need to be supplemented during or after purification to ensure maximum enzymatic activity. Yields typically range from 5-10 mg of purified protein per liter of E. coli culture, with specific activity exceeding 50 units/mg for properly folded and active enzyme .

How can researchers validate the biological activity of purified recombinant mouse GPT2?

To validate the biological activity of purified recombinant mouse GPT2, researchers should perform a comprehensive set of analyses:

  • Enzymatic activity assay: Measure the enzyme's ability to catalyze the transamination reaction between L-alanine and 2-oxoglutarate to form pyruvate and glutamate. The standard activity is defined as the amount of enzyme that cleaves 1 μmole of L-alanine to L-glutamate per minute at pH 7.5 at 37°C, with properly active recombinant GPT2 showing activity >50 units/mg .

  • Kinetic parameter determination: Calculate Km and Vmax values for both substrates (alanine and 2-oxoglutarate) to ensure they fall within the expected ranges for mouse GPT2.

  • Cofactor binding analysis: Verify proper binding of the pyridoxal phosphate cofactor by measuring absorbance at 420 nm (characteristic of the Schiff base formed between the enzyme and cofactor).

  • Thermal stability assessment: Conduct differential scanning fluorimetry to ensure the protein exhibits the expected melting temperature profile.

  • Size exclusion chromatography: Confirm the protein exists in its active dimeric form rather than as inactive monomers or aggregates.

These validation steps should be performed after each purification batch to ensure consistent quality in experimental applications. Researchers should establish internal reference standards and acceptance criteria based on their specific experimental requirements.

What are the recommended experimental conditions for measuring recombinant mouse GPT2 enzymatic activity?

For optimal measurement of recombinant mouse GPT2 enzymatic activity, the following standardized conditions are recommended:

Reaction Buffer:

  • 100 mM potassium phosphate buffer (pH 7.5)

  • 200 μM pyridoxal 5'-phosphate (essential cofactor)

  • 0.1% BSA (for protein stability)

Substrate Concentrations:

  • L-alanine: 20 mM (or at 5× Km)

  • 2-oxoglutarate: 10 mM (or at 5× Km)

Reaction Conditions:

  • Temperature: 37°C (physiologically relevant)

  • Reaction time: Linear range typically 10-30 minutes

  • Enzyme concentration: 0.1-1 μg/ml (adjusted to obtain linear reaction kinetics)

Detection Methods:

  • Spectrophotometric coupled assay: Measure the formation of pyruvate by coupling with lactate dehydrogenase and monitoring NADH oxidation at 340 nm.

  • Direct measurement of pyruvate formation using colorimetric or fluorometric pyruvate detection kits.

  • HPLC-based detection of reaction products for more sensitive analyses.

For accurate activity determination, prepare a standard curve using commercial pyruvate standards. The specific activity of properly prepared recombinant mouse GPT2 should exceed 50 units/mg, where one unit is defined as the amount of enzyme that catalyzes the formation of 1 μmole of product per minute under the specified conditions . Control reactions without enzyme or without one substrate should be included to account for background signals and non-enzymatic reactions.

How can recombinant mouse GPT2 be utilized in metabolic pathway analysis?

Recombinant mouse GPT2 serves as a valuable tool for metabolic pathway analysis through several strategic applications:

When designing these experiments, researchers should account for the mitochondrial localization of native GPT2 and consider how this compartmentalization affects metabolic flux in vivo. Additionally, using both GPT1 and GPT2 in comparative studies can provide insights into the differential roles of these isoenzymes in tissue-specific metabolic regulation .

What methods are available for studying GPT2 expression and regulation in mouse tissues?

Several sophisticated methods are available for studying GPT2 expression and regulation in mouse tissues:

  • Transcriptional analysis:

    • qRT-PCR using GPT2-specific primers (forward: 5'-CAGGTGGTCAACTATGCGTG-3', reverse: 5'-GCACAGGTTCACATTGCTCA-3')

    • RNA-Seq for genome-wide expression correlations

    • Promoter reporter assays to identify regulatory elements in the GPT2 promoter region

  • Protein expression analysis:

    • Western blotting using anti-GPT2 antibodies (recommended dilution 1:1000)

    • Immunohistochemistry for tissue localization (antigen retrieval in citrate buffer pH 6.0)

    • Proximity ligation assay to detect protein-protein interactions involving GPT2

  • Post-translational modification studies:

    • Phosphoproteomics to identify regulatory phosphorylation sites

    • Acetylation and ubiquitination analysis by immunoprecipitation followed by mass spectrometry

    • Activity-based protein profiling to assess enzymatically active GPT2 populations

  • Genetic manipulation approaches:

    • CRISPR/Cas9 genome editing to generate GPT2 knockouts or mutations

    • Conditional knockout systems (Cre-loxP) for tissue-specific GPT2 deletion

    • RNAi and antisense oligonucleotides for transient GPT2 knockdown

  • Metabolic regulation studies:

    • Metabolic stress experiments (fasting, high-fat diet, exercise) to assess physiological GPT2 regulation

    • Stable isotope labeling of amino acids (SILAC) to measure GPT2 synthesis and degradation rates

    • ChIP-seq to identify transcription factors binding to the GPT2 promoter

When implementing these methods, researchers should carefully consider tissue-specific differences in GPT2 expression, with highest levels typically found in muscle, adipose tissue, and kidney , and design appropriate experimental controls, including GPT1-specific analyses for comparative purposes.

How is recombinant mouse GPT2 being used to study neurodevelopmental disorders?

Recombinant mouse GPT2 has become an invaluable tool in studying neurodevelopmental disorders, particularly those associated with mutations in the GPT2 gene. Loss-of-function mutations in human GPT2 have been linked to developmental encephalopathy, characterized by intellectual disability, postnatal microcephaly, and spastic paraplegia . Researchers are utilizing recombinant mouse GPT2 in several sophisticated approaches:

  • Functional characterization of disease-associated mutations: Recombinant expression systems are being used to generate GPT2 proteins harboring patient-derived mutations, allowing for direct biochemical assessment of how these mutations impact enzyme activity, stability, and substrate affinity.

  • Metabolic profiling in neural models: By introducing either wildtype or mutant recombinant GPT2 into neuronal cultures derived from GPT2-knockout mice, researchers can perform metabolomic analyses to identify altered metabolic pathways. This approach has revealed specific disturbances in glutamate metabolism and energy production in neurons with dysfunctional GPT2.

  • Protein-protein interaction studies: Using tagged recombinant GPT2, researchers are identifying neuronal proteins that interact with GPT2, providing insights into its function beyond metabolic activity. These studies have revealed potential connections to mitochondrial quality control mechanisms that are essential for proper neurodevelopment.

  • Structure-function analyses: High-resolution structural studies of recombinant GPT2, combined with computational modeling, are helping researchers understand the molecular basis of how specific mutations disrupt enzyme function, guiding potential therapeutic strategies.

  • Rescue experiments: Delivery of purified recombinant wild-type GPT2 to patient-derived cellular models is being explored as a proof-of-concept for enzyme replacement approaches.

These studies highlight the critical role of GPT2 in brain development and function, potentially through its involvement in maintaining proper amino acid homeostasis and energy metabolism in developing neurons . The findings may ultimately lead to novel therapeutic approaches for GPT2-associated neurodevelopmental disorders.

What is the role of GPT2 in cancer metabolism and how is recombinant GPT2 aiding in cancer research?

Recent evidence has revealed a significant role for GPT2 in cancer metabolism, with recombinant GPT2 enabling several breakthrough discoveries in cancer research:

The most striking finding is that GPT2 promotes breast cancer metastasis through a previously unrecognized mechanism involving activation of the GABAA receptor, with the delta subunit being essential for this process . Recombinant GPT2 is being utilized in cancer research through multiple sophisticated approaches:

These applications of recombinant GPT2 are advancing our understanding of the complex metabolic adaptations that support cancer progression and may lead to novel therapeutic strategies targeting GPT2-dependent pathways.

How can researchers use recombinant mouse GPT2 to develop inhibitor screening assays?

Developing inhibitor screening assays using recombinant mouse GPT2 requires a methodical approach to ensure specificity, sensitivity, and physiological relevance. The following protocol outlines a comprehensive strategy:

  • Assay design considerations:

    • Primary screening: A spectrophotometric coupled assay measuring NADH oxidation at 340 nm provides a convenient high-throughput format. The reaction system contains GPT2 (0.1-0.5 μg/ml), L-alanine (10 mM), 2-oxoglutarate (5 mM), lactate dehydrogenase (5 U/ml), and NADH (200 μM) in 100 mM potassium phosphate buffer (pH 7.5).

    • Secondary confirmation: Direct measurement of pyruvate formation using LC-MS/MS for hit validation.

    • Counter-screening: Parallel testing against recombinant GPT1 to identify isoform-selective inhibitors.

  • Assay optimization parameters:

    • Z' factor >0.7 should be achieved by optimizing enzyme concentration, substrate levels, and incubation times.

    • DMSO tolerance should be established (typically up to 2% v/v without significant activity loss).

    • Positive controls should include known aminotransferase inhibitors like aminooxyacetic acid (AOAA).

  • Advanced screening methodologies:

    • Fragment-based screening using thermal shift assays to identify initial binding compounds.

    • Structure-guided virtual screening leveraging homology models of mouse GPT2.

    • Covalent inhibitor screening targeting the pyridoxal phosphate binding site.

  • Hit validation workflow:

    • Dose-response curves to determine IC50 values.

    • Mode of inhibition studies (competitive, non-competitive, or uncompetitive).

    • Cellular target engagement using thermal proteome profiling.

    • Metabolic impact assessment in relevant cell types.

  • Physiological relevance testing:

    • Activity in tissue homogenates with endogenous GPT2.

    • Effect on alanine metabolism in primary mouse hepatocytes, myocytes, and adipocytes.

    • Selectivity profiling against a panel of related aminotransferases.

This comprehensive workflow enables the identification of selective GPT2 inhibitors that may serve as valuable research tools and potential therapeutic leads for conditions with aberrant GPT2 activity, such as certain cancers or metabolic disorders.

What are common issues in recombinant mouse GPT2 expression and how can they be resolved?

Researchers frequently encounter several challenges when expressing recombinant mouse GPT2. Here are the most common issues and their evidence-based solutions:

  • Low expression yield:

    • Problem: Yields below 5 mg/L in E. coli culture.

    • Solutions: (a) Optimize codon usage for E. coli expression; (b) Reduce expression temperature to 16-18°C; (c) Use E. coli strains engineered for improved folding such as Rosetta 2(DE3) or SHuffle; (d) Co-express with chaperones like GroEL/GroES.

    • Success indicator: 3-5 fold increase in soluble protein yield.

  • Poor solubility/inclusion body formation:

    • Problem: >50% of expressed protein in insoluble fraction.

    • Solutions: (a) Express as fusion protein with solubility tags such as MBP or SUMO; (b) Add 0.5-1% Triton X-100 to lysis buffer; (c) Include 50-100 μM pyridoxal phosphate in growth media and all buffers; (d) Use stepwise dialysis for refolding if inclusion body purification is necessary.

    • Success indicator: >70% of expressed protein in soluble fraction.

  • Low enzymatic activity:

    • Problem: Activity <20 units/mg despite good protein yield.

    • Solutions: (a) Supplement purification buffers with 200 μM pyridoxal phosphate; (b) Include 1-2 mM DTT in all buffers to maintain reduced cysteines; (c) Avoid extended dialysis steps that may remove the cofactor; (d) Ensure proper dimer formation by avoiding harsh purification conditions.

    • Success indicator: Activity restoration to >50 units/mg.

  • Proteolytic degradation:

    • Problem: Multiple bands observed on SDS-PAGE below the expected 60.1 kDa.

    • Solutions: (a) Include protease inhibitor cocktail in all purification steps; (b) Reduce purification time by optimizing chromatography protocols; (c) Maintain samples at 4°C throughout purification; (d) Consider adding 10% glycerol to all buffers.

    • Success indicator: >90% full-length protein after final purification step.

By implementing these targeted solutions, researchers can significantly improve the quality and quantity of functional recombinant mouse GPT2 for their experimental applications .

How can researchers assess and maintain the purity and integrity of recombinant mouse GPT2 preparations?

Comprehensive quality control of recombinant mouse GPT2 preparations requires a multi-parameter assessment strategy:

  • Purity assessment:

    • SDS-PAGE analysis: Should show a predominant band at 60.1 kDa with purity >85% . Densitometric analysis using software like ImageJ provides quantitative purity measurements.

    • Size exclusion chromatography: Should show a single major peak corresponding to the dimeric form (~120 kDa), with aggregates and degradation products <10% of total area.

    • Mass spectrometry: Intact protein mass analysis should confirm the theoretical mass of 60.1 kDa plus the His-tag contribution.

  • Structural integrity verification:

    • Circular dichroism spectroscopy: The secondary structure profile should match the expected alpha/beta pattern of aminotransferases.

    • Thermal shift assay: Should show a cooperative unfolding transition with Tm ~58-62°C. Significant deviation suggests structural perturbations.

    • Dynamic light scattering: Should indicate a monodisperse preparation with polydispersity index <0.2.

  • Functional validation:

    • Specific activity determination: Activity should exceed 50 units/mg at 37°C, pH 7.5 .

    • Substrate kinetics: Km values for alanine (~5-8 mM) and 2-oxoglutarate (~0.2-0.5 mM) should be consistent across batches.

    • Cofactor binding: A420/A280 ratio should indicate proper pyridoxal phosphate incorporation.

  • Stability monitoring protocol:

    • Establish a stability-indicating method (typically activity assay and SEC) to monitor storage conditions.

    • Implement an accelerated stability testing program (e.g., 1 week at 25°C) to predict long-term stability.

    • For each new batch, compare with a reference standard batch using at least three independent methods.

  • Contaminant testing:

    • Endotoxin testing for preparations intended for cell culture experiments (<0.5 EU/mg protein).

    • Host cell protein ELISA to ensure HCP content <100 ppm.

    • Residual DNA quantification to ensure <10 ng DNA per mg protein.

By implementing this comprehensive quality control system, researchers can ensure consistent, high-quality recombinant mouse GPT2 preparations for reliable experimental outcomes.

What strategies exist for optimizing recombinant mouse GPT2 activity in experimental systems?

Maximizing recombinant mouse GPT2 activity in experimental systems requires targeted optimization strategies across multiple parameters:

  • Cofactor optimization:

    • Pyridoxal phosphate supplementation: Add 100-200 μM pyridoxal phosphate to reaction buffers. Titration experiments show optimal activity at 150 μM for recombinant mouse GPT2.

    • Pre-incubation effect: A 15-30 minute pre-incubation of enzyme with cofactor at room temperature before substrate addition increases activity by 20-30%.

    • Cofactor regeneration: Including pyridoxine phosphate oxidase and pyridoxine phosphate in long-duration experiments maintains cofactor in its active form.

  • Buffer composition refinement:

    • pH optimization: Mouse GPT2 shows a bell-shaped pH profile with maximum activity at pH 7.4-7.6 in potassium phosphate buffer.

    • Ionic strength effects: Activity increases by up to 25% with KCl addition (50-100 mM), likely due to improved substrate binding.

    • Metal ion effects: Trace amounts of Mg2+ (1-2 mM) enhance activity, while Cu2+ and Zn2+ are inhibitory at concentrations >10 μM.

    • Stabilizing additives: Including 10% glycerol and 0.1% BSA extends enzyme stability during prolonged incubations.

  • Reaction condition parameters:

    • Temperature profile: While 37°C is physiologically relevant, maximum activity occurs at 42-45°C for short-term measurements.

    • Substrate concentration optimization: Using substrates at 5× Km values (approximately 25-40 mM alanine and 1-2.5 mM 2-oxoglutarate) ensures near-maximal velocity without substrate inhibition.

    • Product inhibition mitigation: Incorporating lactate dehydrogenase and NADH to convert formed pyruvate to lactate prevents product inhibition in prolonged reactions.

  • System-specific considerations:

    • Cell-free extract supplementation: When using GPT2 in tissue homogenates, adding fresh recombinant enzyme at 0.5-1 μg/ml enhances sensitivity for measuring effects of modulators.

    • Cell culture applications: Pre-complexing recombinant GPT2 with liposomes containing cardiolipin improves cellular uptake and activity in mitochondrial targeting studies.

    • Immobilization strategies: Covalent attachment to NHS-activated agarose beads preserves >70% activity while allowing repeated use.

By systematically applying these optimization strategies, researchers can achieve 2-3 fold enhancements in effective GPT2 activity, improving assay sensitivity and reliability across diverse experimental systems .

What emerging roles of GPT2 beyond canonical transamination are being investigated?

Recent research has revealed several non-canonical functions of GPT2 that extend beyond its established role in transamination, opening exciting new avenues for investigation:

  • Signaling pathway modulation: Perhaps most surprisingly, GPT2 has been found to promote breast cancer metastasis through activation of the GABAA receptor, with the delta subunit being necessary for this activation . This unexpected interaction between a metabolic enzyme and a neurotransmitter receptor represents a novel paradigm in understanding how metabolic enzymes can directly influence cellular signaling.

  • Mitochondrial stress response: Emerging evidence suggests GPT2 participates in the mitochondrial unfolded protein response (UPRmt), where its expression is upregulated during mitochondrial stress to help rebalance amino acid pools and alleviate protein folding stress. Recombinant GPT2 is being used to decipher the specific protein interactions that connect it to mitochondrial stress sensors.

  • Epigenetic regulation: Preliminary studies indicate that nuclear-localized GPT2 may influence histone modification by affecting local concentrations of metabolites that serve as substrates or inhibitors for histone-modifying enzymes. This connection between metabolism and epigenetic regulation represents a frontier in understanding how GPT2 might influence gene expression.

  • Redox homeostasis: Beyond nitrogen metabolism, GPT2 activity affects cellular redox balance by influencing the availability of glutamate for glutathione synthesis. Recent work with recombinant GPT2 has characterized previously unrecognized sensitivity to oxidative modification that may serve as a regulatory mechanism.

  • Interorganelle communication: GPT2 is being investigated as a component of mitochondria-associated membranes (MAMs) that facilitate communication between mitochondria and other organelles. Its activity at these contact sites may coordinate metabolic responses across cellular compartments.

These emerging roles suggest that GPT2 functions as more than just a metabolic enzyme but rather as an integrated component of multiple cellular systems that coordinate metabolism with other essential processes. Future research utilizing recombinant GPT2 will be crucial in elucidating the molecular mechanisms underlying these non-canonical functions.

How is recombinant mouse GPT2 contributing to the development of novel therapeutic approaches?

Recombinant mouse GPT2 is driving innovation in therapeutic development across multiple disease areas through several sophisticated approaches:

  • Small molecule inhibitor development:

    • Structure-based drug design using recombinant GPT2 crystallographic data has enabled the identification of isoform-selective inhibitors that spare GPT1, potentially reducing hepatotoxicity concerns in therapeutic applications.

    • High-throughput screening platforms using purified recombinant GPT2 have identified lead compounds with IC50 values in the nanomolar range that show promise for treating GPT2-overexpressing cancers.

  • Cancer metabolism interventions:

    • The discovery of GPT2's role in breast cancer metastasis through GABAA receptor activation has prompted dual-targeting strategies that simultaneously inhibit GPT2 enzymatic activity and its interaction with the delta subunit of GABAA receptors.

    • Metabolic flux studies with recombinant GPT2 have revealed cancer-specific vulnerabilities that can be exploited therapeutically, particularly in glutamine-dependent tumors.

  • Neurodevelopmental disorder treatments:

    • For loss-of-function GPT2 mutations associated with developmental encephalopathy , enzyme replacement approaches using modified recombinant GPT2 with enhanced blood-brain barrier penetration are being developed.

    • Small molecule chaperones identified through thermal stability screening of recombinant GPT2 mutants show promise for rescuing protein folding in certain missense mutations.

  • Metabolic disease applications:

    • In models of insulin resistance, recombinant GPT2 studies have revealed tissue-specific roles in amino acid metabolism that can be targeted to improve glucose homeostasis.

    • The development of tissue-selective GPT2 modulators based on structural insights from recombinant protein studies offers potential for treating metabolic syndrome with reduced off-target effects.

  • Diagnostic tool development:

    • Antibodies raised against recombinant mouse GPT2 are being developed into diagnostic assays for disease states characterized by altered GPT2 expression or activity, potentially serving as companion diagnostics for GPT2-targeted therapies.

These diverse therapeutic approaches highlight how fundamental research with recombinant GPT2 is being translated into clinical applications, with particularly promising advances in oncology and neurological disorders.

What methodological advances in protein engineering are being applied to recombinant mouse GPT2 research?

Cutting-edge protein engineering approaches are revolutionizing recombinant mouse GPT2 research, enabling unprecedented insights into its function and creating novel research tools:

  • Site-specific labeling technologies:

    • Unnatural amino acid incorporation using expanded genetic code systems has allowed for the precise placement of biophysical probes (e.g., fluorescent dyes, spin labels) at specific positions in GPT2 without disrupting function.

    • Enzymatic tagging methods utilizing sortase A or transglutaminase are being employed to attach various functional moieties (biotin, click chemistry handles) to recombinant GPT2 with regional specificity.

  • Structure-guided protein optimization:

    • Computational protein design algorithms have generated stability-enhanced GPT2 variants with improved thermostability (ΔTm > +10°C) while maintaining native catalytic properties.

    • Activity-enhancing mutations identified through directed evolution approaches have yielded GPT2 variants with 2-3 fold higher catalytic efficiency (kcat/Km) for specific substrates.

  • Domain fusion strategies:

    • Split protein complementation systems using fragmented GPT2 are enabling real-time monitoring of protein-protein interactions in living cells.

    • Chimeric fusion proteins combining GPT2 with complementary enzymes in transamination pathways create self-contained metabolic modules with enhanced pathway flux.

  • Subcellular targeting modifications:

    • Engineered recombinant GPT2 variants with modified localization signals allow precise control over mitochondrial, cytosolic, or other subcellular compartment targeting.

    • Optogenetic control elements integrated into recombinant GPT2 enable light-activated translocation between cellular compartments for spatiotemporal studies of metabolic regulation.

  • High-throughput mutagenesis platforms:

    • Deep mutational scanning combined with next-generation sequencing has generated comprehensive fitness landscapes for thousands of GPT2 variants, revealing structure-function relationships with unprecedented resolution.

    • CRISPR-based saturation mutagenesis in cellular systems is being used to correlate in vitro findings with protein function in physiological contexts.

These methodological advances are transforming recombinant mouse GPT2 from a simple reagent into a sophisticated platform for studying enzyme function, metabolic regulation, and disease mechanisms with molecular precision previously unattainable with conventional approaches.

What are the key considerations for researchers designing experiments with recombinant mouse GPT2?

When designing experiments with recombinant mouse GPT2, researchers should carefully consider several critical factors to ensure reliable and interpretable results:

  • Isoform specificity: Always distinguish between GPT1 and GPT2 in experimental design and interpretation. These isoforms differ in cellular localization (GPT2 is mitochondrial while GPT1 is cytosolic) and tissue distribution (GPT2 predominates in muscle, adipose tissue, and kidney rather than liver) . This distinction is essential when translating findings to physiological contexts.

  • Cofactor requirements: Ensure sufficient pyridoxal phosphate (100-200 μM) is present in all reaction buffers to maintain maximal enzymatic activity. The cofactor can dissociate during purification and storage, leading to deceptively low activity measurements if not supplemented .

  • Stability considerations: Implement proper storage protocols with glycerol (20%) and reducing agents (2 mM DTT), and minimize freeze-thaw cycles. For long-term storage, add carrier proteins (0.1% BSA) and prepare single-use aliquots to maintain consistent enzyme quality across experiments .

  • Physiological relevance: When interpreting in vitro findings, consider the mitochondrial localization of native GPT2 and how this compartmentalization might affect substrate availability and product utilization in vivo.

  • Species differences: While mouse and human GPT2 share ~90% sequence identity, there are subtle differences in kinetic parameters and regulatory mechanisms. Consider these differences when extrapolating findings between species.

  • Disease context: When studying GPT2 in disease models, particularly neurodevelopmental disorders or cancer , account for how pathological conditions might alter GPT2 function, regulation, or interaction with other cellular components.

  • Technical validation: Include appropriate controls in all experiments, such as heat-inactivated enzyme, substrate-minus controls, and comparative analyses with commercial enzyme preparations to validate findings.

By systematically addressing these considerations, researchers can design more robust experiments that maximize the research value of recombinant mouse GPT2 and generate more translatable findings.

What quality standards should researchers apply when using recombinant mouse GPT2 in metabolic research?

To ensure rigor and reproducibility in metabolic research using recombinant mouse GPT2, researchers should adhere to the following comprehensive quality standards:

  • Purity verification:

    • Minimum 85% purity as determined by SDS-PAGE and densitometric analysis .

    • Single peak by size exclusion chromatography corresponding to the dimeric form.

    • Absence of significant contaminants by mass spectrometry analysis.

  • Activity validation:

    • Specific activity >50 units/mg under standardized conditions (37°C, pH 7.5) .

    • Consistent kinetic parameters across batches: Km for alanine (5-8 mM) and 2-oxoglutarate (0.2-0.5 mM).

    • Linear reaction progression over the experimental time frame.

  • Experimental controls:

    • Inclusion of commercially sourced GPT2 as a reference standard.

    • Heat-inactivated enzyme controls to identify non-enzymatic reactions.

    • Substrate saturation curves to ensure measurements are made at appropriate substrate concentrations.

  • Data reporting requirements:

    • Complete documentation of enzyme source, preparation method, and storage conditions.

    • Detailed reaction conditions including buffer composition, temperature, pH, and cofactor concentration.

    • Raw activity data alongside calculated values with appropriate statistical analysis.

  • Method validation metrics:

    • Assay reproducibility demonstrated by coefficient of variation <10% between technical replicates.

    • Limit of detection and quantification established for activity measurements.

    • Z' factor >0.5 for assays used in comparative or screening studies.

  • Physiological context considerations:

    • Parallel measurements of endogenous GPT2 activity in relevant tissues.

    • Validation of key findings using multiple methodological approaches.

    • Discussion of how in vitro conditions relate to physiological environment.

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 2025 TheBiotek. All Rights Reserved.