GOT2 Human, Active

Glutamic-Oxaloacetic Transaminase 2 Human Recombinant, Active
Shipped with Ice Packs
In Stock

Description

Introduction to GOT2 Human, Active

GOT2 (Glutamic-Oxaloacetic Transaminase 2), also known as mitochondrial aspartate aminotransferase, is a pyridoxal phosphate (PLP)-dependent enzyme encoded by the GOT2 gene in humans. It plays a pivotal role in amino acid metabolism, the malate-aspartate shuttle, and the tricarboxylic acid (TCA) cycle . The "active" form refers to its catalytically functional state, typically purified or recombinant, used for biochemical and therapeutic research.

Key Features:

  • Gene ID: 2806 (HGNC: 4433)

  • Protein Length: 430 amino acids

  • Molecular Weight: ~47.5 kDa

  • Structure: Homodimer with PLP cofactors bound to lysine residues

  • Isoforms: Two splice variants (isoforms 1 and 2)

SpeciesLength (aa)Mass (Da)UniProt ID
Human43047,518P00505
Mouse43047,411P05202
Rat43047,314P00506
Data derived from Sino Biological .

Catalytic Function:

GOT2 catalyzes the reversible transamination of L-aspartate and α-ketoglutarate to oxaloacetate and L-glutamate, critical for:

  • Malate-Aspartate Shuttle: Maintaining NAD+/NADH balance between mitochondria and cytosol .

  • Amino Acid Metabolism: Linking the urea and TCA cycles .

  • Redox Homeostasis: Mitigating reductive stress in pancreatic cancer cells .

Activity Assay:

  • Standard assays measure conversion rates of α-ketoglutarate to L-glutamate (activity >20 units/mg at pH 8.0, 25°C) .

Cancer Metabolism:

  • Pancreatic Ductal Adenocarcinoma (PDAC):

    • GOT2 sustains tumor growth by suppressing senescence via p27-dependent pathways .

    • Regulates immune evasion by promoting PPARδ transcriptional activity, restricting CD4+/CD8+ T-cell infiltration .

  • Metabolic Plasticity: Cancer cells compensate for GOT2 inhibition via microenvironmental pyruvate uptake .

Neurological Disorders:

  • Developmental and Epileptic Encephalopathy 82 (DEE82): Linked to GOT2 mutations .

Technical Considerations for Experimental Use

  • Subcellular Localization: Primarily mitochondrial, but nuclear pools require specific detection methods .

  • Inhibitors: Aminooxyacetic acid (AOA) reduces complex II respiration in muscle mitochondria .

  • Redundancy: GOT1 (cytosolic isoform) may compensate for GOT2 loss in certain contexts .

Product Specs

Introduction
GOT2, also known as Mitochondrial Aspartate Aminotransferase (mAST), is a key enzyme in amino acid metabolism. It exists in both cytoplasmic (GOT1) and mitochondrial (GOT2) forms, playing crucial roles in the urea cycle and the tricarboxylic acid (TCA) cycle. Both GOT1 and GOT2 are homodimeric enzymes and share a high degree of homology.
Description
This product consists of recombinant human GOT2, produced in E. coli. It is a single, non-glycosylated polypeptide chain encompassing amino acids 30 to 430, resulting in a molecular weight of 47 kDa. A 23 amino acid histidine tag is fused to the N-terminus to facilitate purification, which is achieved using proprietary chromatographic methods.
Physical Appearance
The product is a sterile, colorless solution.
Formulation
The GOT2 is supplied in a solution at a concentration of 0.5 mg/ml. The formulation buffer consists of 20 mM Tris-HCl (pH 8.0), 0.15 M NaCl, 10% glycerol, and 1 mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure optimal stability during long-term storage, adding a carrier protein such as HSA or BSA (0.1%) is advisable. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of the GOT2 is greater than 95%, as determined by SDS-PAGE analysis.
Biological Activity
The specific activity of GOT2 is greater than 60 units/mg. One unit of activity is defined as the amount of enzyme required to catalyze the conversion of 1 µmol of α-ketoglutarate to L-Glutamate per minute at a pH of 8.0 and a temperature of 25°C.
Synonyms
EC 2.6.1.1, Aspartate aminotransferase 1, Transaminase A, GIG18.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSSSWWTHV EMGPPDPILG VTEAFKRDTN SKKMNLGVGA YRDDNGKPYV LPSVRKAEAQ IAAKNLDKEY LPIGGLAEFC KASAELALGE NSEVLKSGRF VTVQTISGTG ALRIGASFLQ RFFKFSRDVF LPKPTWGNHT PIFRDAGMQL QGYRYYDPKT CGFDFTGAVE DISKIPEQSV LLLHACAHNP TGVDPRPEQW KEIATVVKKR NLFAFFDMAY QGFASGDGDK DAWAVRHFIE QGINVCLCQS YAKNMGLYGE RVGAFTMVCK DADEAKRVES QLKILIRPMY SNPPLNGARI AAAILNTPDL RKQWLQEVKV MADRIIGMRT QLVSNLKKEG STHNWQHITD QIGMFCFTGL KPEQVERLIK EFSIYMTKDG RISVAGVTSS NVGYLAHAIH QVTK.

Q&A

What is the molecular structure of GOT2 and how does it relate to its enzymatic function?

GOT2 is a homodimeric enzyme containing two identical subunits with overlapping regions. The enzyme's structural organization features helices forming the top and sides, while the bottom consists of beta sheets and extended hairpin loops. Each subunit can be divided into four distinct parts: a large domain that binds pyridoxal-P (the cofactor essential for transamination reactions), a small domain, an NH2-terminal arm, and a bridge spanning the two domains (formed by residues 48-75 and 301-358) .

The pyridoxal phosphate binding site in the large domain is critical for GOT2's catalytic activity, enabling the transamination reactions that convert oxaloacetate to aspartate. This structural arrangement facilitates efficient substrate binding and catalysis. The homodimeric nature provides stability and may enable cooperative interactions between subunits during catalysis. GOT2's genetic structure reflects its importance as a housekeeping gene, with its 5' regulatory regions lacking a TATA box, and the gene being located on chromosome 16q21 with 10 exons .

Understanding this structure-function relationship is essential for designing experiments to investigate GOT2's role in various metabolic pathways and for developing potential modulators of its activity.

How does GOT2 contribute to cellular energy metabolism through the malate-aspartate shuttle?

GOT2 plays a critical role in the malate-aspartate shuttle, which is essential for transferring reducing equivalents (NADH) from the cytosol to the mitochondria. This process is necessary because the inner mitochondrial membrane is impermeable to NADH, requiring an indirect transfer mechanism to support mitochondrial energy production .

The specific function of GOT2 in this shuttle involves:

  • Inside the mitochondria, GOT2 catalyzes the conversion of oxaloacetate to aspartate through transamination with glutamate, producing α-ketoglutarate

  • The aspartate then exits the mitochondria via specific transporters

  • In the cytosol, GOT1 (the cytosolic counterpart) converts aspartate back to oxaloacetate, completing part of the shuttle cycle

This shuttle is particularly important for maintaining high rates of glycolysis, as it ensures rapid regeneration of NAD+ in the cytosol. Research has shown that inhibiting the malate-aspartate shuttle impairs glycolysis and decreases cell proliferation in cancer cell lines, highlighting GOT2's importance in cellular energy production .

The shuttling process contributes to cellular energy production by:

  • Enabling cytosolic NADH to contribute to mitochondrial ATP production

  • Supporting continued high rates of glycolysis

  • Integrating with the TCA cycle and oxidative phosphorylation

What distinguishes GOT2 from GOT1, and why is this distinction important for research?

While GOT1 and GOT2 catalyze similar transamination reactions, several critical differences impact their research applications:

CharacteristicGOT1GOT2
Cellular localizationCytoplasmicMitochondrial (inner membrane)
Primary metabolic roleCytosolic amino acid metabolismMalate-aspartate shuttle, mitochondrial metabolism
Gene locationDifferent chromosomeLocated on 16q21
Response to cellular stressDifferent expression patternsExpression often altered in cancer and metabolic disorders

These distinctions are important for researchers because:

  • Experimental designs must account for the subcellular localization of GOT2 when preparing samples and interpreting results. Proper isolation of mitochondrial fractions is essential for accurately measuring GOT2 activity without GOT1 contamination.

  • When studying the malate-aspartate shuttle, researchers must differentiate between the contributions of GOT1 and GOT2, as they catalyze opposite reactions in different cellular compartments.

  • Cancer studies should consider the distinct roles of GOT1 and GOT2 in metabolic reprogramming, as they may be differentially regulated in various cancer types and contribute differently to tumor metabolism .

  • Therapeutic approaches targeting GOT2 should be specific enough to avoid affecting GOT1, which could lead to unintended metabolic consequences.

How is GOT2 expression altered in different cancer types, and what are the clinical implications?

GOT2 expression patterns vary significantly across cancer types, with important clinical implications:

In Hepatocellular Carcinoma (HCC): GOT2 is significantly downregulated in tumor tissues compared to normal liver tissue. Low expression of GOT2 is associated with advanced progression and poor prognosis in HCC patients. Mechanistically, this downregulation promotes tumor growth and metastasis through metabolic reprogramming .

The clinical implications of altered GOT2 expression include:

  • Prognostic value: Low GOT2 expression consistently correlates with worse clinical outcomes in multiple cancer types, suggesting its potential as a prognostic biomarker.

  • Therapeutic vulnerability: Cancer cells with low GOT2 expression become highly dependent on glutamine metabolism and show increased sensitivity to glutaminase inhibitors like CB-839 both in vitro and in vivo, revealing a potential precision medicine approach .

  • Metastatic potential: Loss of GOT2 promotes both hematogenous and intrahepatic metastasis in HCC mouse models, indicating its role in controlling cancer cell invasiveness .

  • Immune microenvironment: GOT2 expression has been linked to immune cell infiltration in tumors, suggesting that it may influence anti-tumor immunity and potentially impact immunotherapy response .

What mechanisms regulate GOT2 expression in cancer cells, and how can researchers investigate these regulatory pathways?

Multiple regulatory mechanisms control GOT2 expression in cancer cells:

DNA Methylation: A primary epigenetic mechanism regulating GOT2 expression is DNA methylation in promoter-related CpG islands. In KIRC, downregulation of GOT2 is driven by hypermethylation of these regulatory regions . To investigate this mechanism, researchers should:

  • Perform bisulfite sequencing of the GOT2 promoter region

  • Use methylation-specific PCR to analyze key regulatory regions

  • Apply methylation inhibitors (e.g., 5-azacytidine) to determine if GOT2 expression can be restored

Transcriptional Regulation: The GOT2 gene has regulatory elements resembling those of typical housekeeping genes, lacking a TATA box . Researchers can investigate transcriptional regulation through:

  • Chromatin immunoprecipitation (ChIP) to identify transcription factors binding to the GOT2 promoter

  • Luciferase reporter assays with wild-type and mutated promoter constructs

  • Analysis of transcription factor expression in relation to GOT2 levels across cancer datasets

Post-transcriptional Regulation: microRNAs and RNA-binding proteins may regulate GOT2 mRNA stability and translation. Investigation approaches include:

  • microRNA prediction tools followed by validation with luciferase assays

  • RNA immunoprecipitation to identify RNA-binding proteins interacting with GOT2 mRNA

  • Analysis of GOT2 mRNA half-life in different cancer contexts

Post-translational Regulation: GOT2 protein stability and activity may be regulated by post-translational modifications. Research methods include:

  • Western blotting with phospho-specific antibodies

  • Mass spectrometry to identify modifications

  • In vitro kinase assays to determine regulatory enzymes

These regulatory mechanisms provide potential therapeutic targets for modulating GOT2 expression and activity in cancer treatment strategies.

How does GOT2 silencing affect cancer cell metabolism and what are the therapeutic implications?

GOT2 silencing leads to complex metabolic reprogramming in cancer cells with significant therapeutic implications:

Metabolic Effects:

  • Enhanced glutaminolysis: Silencing of GOT2 promotes increased glutamine metabolism in cancer cells .

  • Increased nucleotide synthesis: GOT2 knockdown redirects glutamine-derived carbon toward nucleotide production to support rapid cell proliferation .

  • Elevated glutathione synthesis: Loss of GOT2 enhances glutathione production, strengthening the cellular antioxidant system and promoting cancer cell survival under oxidative stress .

  • PI3K/AKT/mTOR pathway activation: GOT2 silencing activates this oncogenic signaling cascade, contributing to cancer progression .

Experimental Approaches to Study These Effects:

  • Stable isotope tracing with labeled glutamine (13C, 15N) to track metabolic flux

  • Targeted metabolomics to quantify changes in key metabolites

  • Measurements of cellular redox status (GSH/GSSG ratios)

  • Western blotting for activation markers of the PI3K/AKT/mTOR pathway

Therapeutic Implications:
The most significant therapeutic finding is that cancer cells with low GOT2 expression become highly dependent on glutamine metabolism. This metabolic vulnerability creates an opportunity for precision medicine approaches using glutaminase inhibitors such as CB-839, which have shown efficacy against GOT2-low HCC both in vitro and in vivo .

Potential therapeutic strategies include:

  • Using GOT2 expression as a biomarker to identify patients likely to respond to glutaminase inhibitors

  • Developing combination therapies targeting both glutamine metabolism and the PI3K/AKT/mTOR pathway

  • Exploring synthetic lethal interactions with other metabolic pathways in GOT2-low cancers

This research highlights how metabolic adaptations driven by altered GOT2 expression can create targetable dependencies in cancer cells, advancing precision medicine approaches.

What methodological approaches should be used to investigate GOT2's role in tumor progression?

Investigating GOT2's role in tumor progression requires a comprehensive set of methodological approaches:

In Vitro Cell Models:

  • Expression modulation: Generate stable cell lines with GOT2 knockdown (shRNA, CRISPR-Cas9) or overexpression to study effects on cancer phenotypes.

  • Proliferation assays: Measure cell growth using MTT/XTT assays, BrdU incorporation, or real-time cell analysis systems.

  • Migration and invasion assays: Employ wound healing, transwell migration, and Matrigel invasion assays to assess metastatic potential.

  • 3D culture models: Use spheroids or organoids to better recapitulate tumor architecture and microenvironment effects.

Metabolic Analysis:

  • Seahorse analysis: Measure oxygen consumption rate and extracellular acidification rate to assess mitochondrial function and glycolytic activity.

  • Metabolomics: Employ targeted or untargeted approaches to identify metabolic changes.

  • Isotope tracing: Use 13C-labeled glutamine, glucose, or other nutrients to track metabolic flux.

  • Glutathione and ROS measurement: Assess redox status using fluorescent probes and biochemical assays.

In Vivo Models:

  • Xenograft models: Implant GOT2-modified cancer cells subcutaneously or orthotopically in immunocompromised mice.

  • Metastasis models: Use tail vein injection or intrasplenic injection models to assess metastatic potential.

  • Patient-derived xenografts: Evaluate GOT2 expression and function in more clinically relevant models.

  • Transgenic mouse models: Consider inducible, tissue-specific GOT2 knockout or knockin models for developmental studies.

Clinical Correlation:

  • Tissue microarrays: Analyze GOT2 protein expression by immunohistochemistry in patient samples .

  • Analysis of public datasets: Mine TCGA, GEO, and other databases for correlations between GOT2 expression and clinical outcomes.

  • Single-cell RNA-seq: Investigate cell-type specific GOT2 expression within the tumor microenvironment.

  • Liquid biopsy approaches: Explore potential for detecting GOT2 in circulation as a biomarker.

These methodologies should be selected based on specific research questions and integrated to provide comprehensive insights into GOT2's role in tumor progression.

What are the optimal protocols for measuring GOT2 enzymatic activity in research samples?

Measuring GOT2 enzymatic activity requires careful experimental design to ensure specificity and reliability:

Sample Preparation:

  • Mitochondrial Isolation: For specific GOT2 activity measurement (distinct from cytosolic GOT1), proper mitochondrial isolation is critical.

    • Use differential centrifugation or commercial mitochondrial isolation kits

    • Verify mitochondrial purity using Western blotting for compartment-specific markers

    • Maintain samples at 4°C throughout preparation to preserve enzymatic activity

  • Tissue Homogenization:

    • Use appropriate buffers containing protease inhibitors

    • Homogenize tissues using mechanical disruption (Dounce homogenizer or bead-beating)

    • Standardize protein concentration (typically 0.5-2 mg/ml) for comparative analysis

Enzyme Activity Assay Methods:

  • Coupled Spectrophotometric Assay (Gold Standard):

    • Principle: Couple GOT2 reaction with malate dehydrogenase, monitoring NADH oxidation at 340 nm

    • Reaction mix: Aspartate, α-ketoglutarate, NADH, malate dehydrogenase in appropriate buffer

    • Calculate activity from the linear decrease in absorbance using the extinction coefficient of NADH

  • Colorimetric Assays:

    • Commercial kits offer simplified detection with colorimetric endpoints

    • Detection ranges typically span 0.78-50 U/L for mitochondrial GOT2

    • Follow manufacturer protocols precisely for optimal results

Quality Control Measures:

  • Include positive controls (purified GOT2 enzyme or known high-expression samples)

  • Include negative controls (heat-inactivated samples or specific inhibitors)

  • Run technical replicates (minimum triplicate measurements)

  • Establish linear range and optimize enzyme concentration

Data Analysis:

  • Express activity in appropriate units (U/L or U/mg protein)

  • Normalize to mitochondrial content when comparing across different samples

  • Account for potential temperature and pH variations during measurement

  • Use statistical methods appropriate for enzymatic data (non-parametric tests if distribution is non-normal)

These methodological considerations ensure accurate measurement of GOT2 activity for research applications.

What are the most sensitive and specific methods for detecting GOT2 protein levels in human samples?

Several techniques are available for detecting GOT2 protein levels in human samples, each with specific advantages:

Enzyme-Linked Immunosorbent Assay (ELISA):

  • Sandwich ELISA provides high sensitivity and specificity for GOT2 detection in various sample types:

    • Detection range: 1.563-50 ng/mL (typical for human GOT2)

    • Minimum detection limit: Approximately 1.563 ng/mL

    • Sample types: Human plasma, serum, saliva, CSF, cell culture, and tissue homogenates

  • Protocol Optimization:

    • Pre-coat plates with a polyclonal antibody specific for human GOT2

    • Use biotinylated detection antibodies with streptavidin-peroxidase conjugates

    • Implement a standard 4-hour protocol: 2h sample incubation, 1h antibody incubation, 30min conjugate incubation

    • Include high-quality standards for accurate quantification

Immunohistochemistry (IHC):

  • Tissue Preparation:

    • Formalin-fixed paraffin-embedded or frozen sections (4-5 μm thickness)

    • Appropriate antigen retrieval methods (typically heat-induced in citrate buffer)

  • Staining and Analysis:

    • Use validated antibodies for human GOT2 (e.g., HPA018139 as used in Human Protein Atlas)

    • Score based on staining intensity (negative, weak, moderate, or strong) and fraction of stained cells (<25%, 25-75%, or >75%)

    • Require manual annotation by specialists with verification for reliable results

Western Blotting:

  • Sample Preparation:

    • For mitochondrial GOT2, ensure proper subcellular fractionation

    • Use appropriate lysis buffers with protease inhibitors

    • Load equal amounts of protein (15-30 μg) per lane

  • Detection Optimization:

    • Use highly specific primary antibodies

    • Optimize blocking conditions to minimize background

    • Employ enhanced chemiluminescence or fluorescent detection systems

    • Quantify relative to appropriate loading controls

Mass Spectrometry:

  • Sample Processing:

    • Employ efficient protein extraction and digestion protocols

    • Consider enrichment strategies for low-abundance samples

  • Analysis Approaches:

    • Targeted approaches using multiple reaction monitoring (MRM) for quantification

    • Data-dependent acquisition for discovery-based investigations

    • Label-free or isotope-labeled quantification methods

These techniques can be selected based on sample availability, required sensitivity, and specific research questions. For clinical investigations, standardized protocols with appropriate quality controls are essential for reliable results.

What controls and experimental design considerations are critical for GOT2 knockdown studies?

GOT2 knockdown studies require careful experimental design and appropriate controls to ensure valid and interpretable results:

Knockdown Method Selection:

  • siRNA for transient knockdown (3-7 days):

    • Design 3-4 independent siRNA sequences targeting different regions of GOT2 mRNA

    • Test each individually to identify the most effective sequence

    • Use lowest effective concentration to minimize off-target effects

    • Optimal for acute phenotype assessment

  • shRNA for stable knockdown:

    • Use inducible systems (e.g., doxycycline-inducible) to control knockdown timing

    • Generate multiple clones and pool to minimize clonal effects

    • Verify integration site number to avoid multiple insertion effects

    • Suitable for long-term studies including animal models

  • CRISPR-Cas9 for genetic knockout:

    • Design multiple guide RNAs targeting early exons

    • Verify editing by sequencing

    • Generate heterozygous models if homozygous deletion is lethal

    • Create rescue lines re-expressing GOT2 to confirm specificity

Essential Controls:

  • Negative Controls:

    • Non-targeting siRNA/shRNA with identical backbone and delivery method

    • Empty vector controls for expression constructs

    • Cells treated with transfection/transduction reagents alone

    • CRISPR controls with non-targeting guide RNAs

  • Positive Controls:

    • siRNA targeting a gene with well-characterized knockdown phenotype

    • Known modulators of glutamine metabolism pathways

  • Rescue Experiments:

    • Re-expression of siRNA-resistant GOT2 cDNA

    • Expression of enzymatically inactive GOT2 mutants to distinguish catalytic vs. structural roles

Validation Requirements:

  • Transcript Level Validation:

    • qRT-PCR with validated primer pairs

    • Assess potential compensatory changes in related genes (e.g., GOT1)

  • Protein Level Validation:

    • Western blotting with specific antibodies

    • Immunofluorescence to verify subcellular localization changes

  • Functional Validation:

    • Enzymatic activity assays to confirm functional knockdown

    • Metabolic assays to detect expected pathway alterations

Experimental Design Considerations:

  • Cell Line Selection:

    • Consider baseline GOT2 expression levels

    • Use multiple cell lines to ensure robustness of findings

    • Include normal cell counterparts to assess cancer-specific effects

  • Timing of Analysis:

    • Determine optimal time points based on GOT2 protein half-life

    • Include time course experiments to distinguish direct vs. indirect effects

  • Phenotypic Assessment:

    • Proliferation: MTT/XTT, BrdU, colony formation assays

    • Migration/Invasion: Wound healing, transwell assays

    • Metabolism: Glutamine consumption, glutamate production, metabolite profiling

These detailed controls and considerations are critical for generating reliable and reproducible results in GOT2 knockdown studies .

How can researchers effectively quantify changes in glutamine metabolism following GOT2 modulation?

Quantifying changes in glutamine metabolism following GOT2 modulation requires a comprehensive approach combining multiple analytical techniques:

Stable Isotope Tracing:

  • 13C-Glutamine Tracing:

    • Use [U-13C]glutamine or specifically labeled glutamine isotopes (e.g., [1-13C]glutamine)

    • Trace labeled carbon incorporation into downstream metabolites (glutamate, α-ketoglutarate, TCA cycle intermediates)

    • Analyze by GC-MS or LC-MS to determine flux through different metabolic pathways

    • Calculate fractional contribution to determine pathway preferences

  • 15N-Glutamine Tracing:

    • Track nitrogen incorporation into amino acids and nucleotides

    • Particularly useful for assessing transamination reactions governed by GOT2

    • Determine relative activities of competing nitrogen assimilation pathways

Metabolite Quantification:

  • Targeted Metabolomics:

    • Measure levels of glutamine, glutamate, aspartate, and related metabolites

    • Quantify nucleotide pools (particularly important as GOT2 silencing enhances nucleotide synthesis)

    • Assess glutathione levels to evaluate antioxidant system changes

    • Use internal standards for accurate quantification

  • Untargeted Metabolomics:

    • Provides comprehensive metabolic profiling to identify unexpected pathway alterations

    • Requires sophisticated bioinformatic analysis for pattern recognition

    • Useful for generating new hypotheses about metabolic rewiring

Enzymatic Activity Assays:

  • GOT2 Activity:

    • Confirm functional consequences of genetic modulation

    • Use coupled spectrophotometric assays as described in FAQ 4.1

  • Related Enzyme Activities:

    • Glutaminase activity (critical as GOT2-low cells become sensitive to glutaminase inhibitors)

    • Malate dehydrogenase (partner enzyme in the malate-aspartate shuttle)

    • Glutathione synthesis enzymes

Functional Metabolic Assays:

  • Glutamine Dependency:

    • Cell viability in glutamine-depleted media

    • Rescue experiments with glutamine metabolic intermediates

    • Dose-response curves with glutaminase inhibitors (e.g., CB-839)

  • Bioenergetic Analysis:

    • Seahorse XF analysis to measure oxygen consumption rate (OCR) and extracellular acidification rate (ECAR)

    • Mitochondrial stress test with inhibitors to assess respiratory capacity

    • ATP production assays to quantify energy generation

  • Redox Status Assessment:

    • Measure GSH/GSSG ratios to evaluate antioxidant capacity

    • Assess ROS levels using fluorescent probes

    • Measure NADH/NAD+ ratios to evaluate redox balance

These complementary approaches provide a comprehensive view of glutamine metabolic reprogramming following GOT2 modulation, essential for understanding the metabolic vulnerabilities that could be therapeutically targeted .

How does GOT2 interact with oncogenic signaling pathways like PI3K/AKT/mTOR?

GOT2 has a complex relationship with oncogenic signaling pathways, particularly the PI3K/AKT/mTOR pathway, with bidirectional interactions that influence cancer progression:

Mechanisms of Interaction:

  • Metabolic Signaling Integration:

    • GOT2 silencing enhances glutaminolysis, altering cellular energy status and redox balance

    • These metabolic changes can activate mTOR signaling, which is sensitive to cellular nutrient and energy status

    • Enhanced glutathione synthesis resulting from GOT2 silencing protects cancer cells from oxidative stress, further supporting PI3K/AKT/mTOR pathway activation

  • Direct Pathway Activation:

    • Research in hepatocellular carcinoma demonstrates that silencing of GOT2 activates the PI3K/AKT/mTOR pathway, promoting proliferation, migration, and invasion

    • This activation appears to be mediated through metabolic reprogramming, particularly involving altered glutamine metabolism

Experimental Approaches to Study These Interactions:

  • Signaling Analysis:

    • Western blotting for phosphorylated forms of key pathway components (p-AKT, p-mTOR, p-S6K, p-4EBP1)

    • Immunoprecipitation to identify potential protein-protein interactions

    • Pharmacological inhibition of PI3K/AKT/mTOR pathway to determine metabolic consequences

  • Metabolic Intervention Studies:

    • Determine if glutaminase inhibitors (e.g., CB-839) affect PI3K/AKT/mTOR signaling

    • Test whether PI3K/AKT/mTOR inhibitors modulate GOT2 expression or activity

    • Investigate combinatorial approaches targeting both metabolic and signaling nodes

  • In Vivo Validation:

    • Generate xenograft models with GOT2 knockdown and analyze tumors for pathway activation

    • Test combination therapies targeting both GOT2-related metabolism and PI3K/AKT/mTOR signaling

    • Use patient-derived xenografts to validate clinical relevance

Therapeutic Implications:

  • Dual targeting of GOT2 and the PI3K/AKT/mTOR pathway may represent a synergistic approach for cancer treatment

  • In tumors with low GOT2 expression, inhibitors of the PI3K/AKT/mTOR pathway might be particularly effective

  • Metabolic profiling might help identify patients most likely to benefit from such combination approaches

These interactions highlight the complex interplay between metabolic enzymes like GOT2 and major oncogenic signaling pathways, suggesting potential for innovative therapeutic strategies .

What is the relationship between GOT2 expression and tumor microenvironment?

The relationship between GOT2 expression and the tumor microenvironment represents an emerging area of research with important implications for cancer biology and therapy:

Immune Cell Infiltration:

  • Research has suggested that GOT2 expression influences immune cell infiltration in tumors, with potential implications for anti-tumor immunity .

  • Low GOT2 expression may create metabolic conditions in the tumor microenvironment that affect immune cell function and recruitment.

  • Methodological approaches to investigate this relationship include:

    • Multiplex immunohistochemistry to characterize immune cell populations in tumors with varying GOT2 expression

    • Flow cytometry analysis of tumor-infiltrating lymphocytes

    • Single-cell RNA sequencing to assess cell type-specific gene expression profiles

    • Correlation analysis between GOT2 expression and immune signature genes in public datasets

Metabolic Competition:

  • GOT2-low cancer cells show enhanced glutamine metabolism , potentially creating metabolic competition with T cells, which also rely heavily on glutamine.

  • This metabolic competition may contribute to immunosuppression in the tumor microenvironment.

  • Experimental approaches to study this phenomenon include:

    • Co-culture systems with cancer cells and immune cells

    • Metabolite tracing in complex tumor models

    • Analysis of metabolite levels in different regions of tumors using imaging mass spectrometry

Hypoxia and Nutrient Stress:

  • GOT2 function may be particularly important under stress conditions common in the tumor microenvironment, such as hypoxia or nutrient limitation.

  • Changes in GOT2 expression may represent adaptive responses to these microenvironmental stresses.

  • Research methods to explore these relationships include:

    • Culturing cells under hypoxic conditions or nutrient-limited media

    • Using hypoxia-inducible factor (HIF) modulators to mimic hypoxic signaling

    • In vivo models with varying degrees of tumor hypoxia

Stromal Interactions:

  • GOT2 expression in cancer-associated fibroblasts or other stromal cells may influence metabolic symbiosis within the tumor.

  • Metabolite exchange between compartments could be affected by GOT2 activity levels.

  • Investigation approaches include:

    • Laser capture microdissection to analyze compartment-specific gene expression

    • Co-culture models with cancer cells and stromal cells

    • Conditional knockout of GOT2 in specific cell types in vivo

Understanding these complex relationships between GOT2 and the tumor microenvironment could reveal new therapeutic opportunities, potentially combining metabolic targeting with immunotherapy approaches.

What are the mechanisms and implications of epigenetic regulation of GOT2 in cancer?

Epigenetic regulation of GOT2 is emerging as a critical mechanism controlling its expression in cancer, with important biological and clinical implications:

DNA Methylation Mechanisms:

  • GOT2 downregulation is driven by DNA methylation in promoter-related CpG islands in certain cancers, particularly kidney renal clear cell carcinoma (KIRC) .

  • This epigenetic silencing represents a key mechanism by which cancer cells modulate metabolic pathways to support their growth and survival.

  • The methodological approaches to study this mechanism include:

    • Bisulfite sequencing to quantify methylation at specific CpG sites

    • Methylation-specific PCR for targeted analysis of key regulatory regions

    • Treatment with DNA methyltransferase inhibitors (e.g., 5-azacytidine) to determine if GOT2 expression can be restored

    • Correlation analysis between methylation status and expression levels across patient samples

Histone Modifications:

  • In addition to DNA methylation, histone modifications likely play important roles in regulating GOT2 expression.

  • Active chromatin marks (e.g., H3K4me3) or repressive marks (e.g., H3K27me3) at the GOT2 locus can influence its transcriptional activity.

  • Research approaches include:

    • Chromatin immunoprecipitation (ChIP) assays for key histone marks

    • ChIP-seq to map genome-wide distribution of histone modifications

    • Analysis of histone-modifying enzyme expression in relation to GOT2 levels

Functional Consequences:

  • Epigenetic silencing of GOT2 contributes to metabolic reprogramming in cancer cells, including enhanced glutaminolysis and nucleotide synthesis .

  • This metabolic shift supports increased proliferation and creates vulnerabilities that can be therapeutically targeted.

  • Experimental approaches to assess these consequences include:

    • Restoration of GOT2 expression in low-expressing tumors to determine phenotypic effects

    • Metabolic profiling before and after epigenetic drug treatment

    • Sensitivity testing to glutaminase inhibitors in cells with epigenetically silenced GOT2

Clinical Implications:

  • Epigenetic modifications of GOT2 could serve as biomarkers for patient stratification and treatment selection.

  • DNA methylation patterns are stable and can be detected in liquid biopsies, offering potential for non-invasive monitoring.

  • Epigenetic therapies (DNMT inhibitors, HDAC inhibitors) may restore GOT2 expression and potentially sensitize tumors to conventional therapies.

  • Combined approaches targeting both epigenetic mechanisms and resulting metabolic vulnerabilities represent promising therapeutic strategies.

Understanding these epigenetic mechanisms offers insights into cancer development and progression while potentially revealing new approaches for precision medicine strategies targeting GOT2-related metabolic pathways .

How can GOT2-dependent metabolic vulnerabilities be exploited for cancer therapy?

The discovery that GOT2 downregulation creates specific metabolic vulnerabilities offers promising opportunities for targeted cancer therapy:

Glutaminase Inhibition:

  • Research has demonstrated that HCC with low expression of GOT2 becomes highly dependent on glutamine metabolism, creating a therapeutic vulnerability .

  • GOT2-low cancers show enhanced sensitivity to glutaminase inhibitors like CB-839 both in vitro and in vivo .

  • Methodological approaches for exploiting this vulnerability include:

    • Cell viability assays with dose ranges of glutaminase inhibitors in GOT2-high versus GOT2-low cells

    • In vivo efficacy studies using patient-derived xenografts with varying GOT2 expression

    • Combination studies with standard of care therapies to identify synergistic interactions

    • Metabolic flux analysis to confirm mechanism of action and identify resistance mechanisms

Biomarker Development:

  • GOT2 expression could serve as a predictive biomarker for response to metabolic-targeted therapies.

  • Development approaches include:

    • Immunohistochemical staining protocols for clinical samples using validated antibodies

    • ELISA-based quantification in liquid biopsies

    • Gene expression signatures that incorporate GOT2 and related metabolic enzymes

    • Metabolite profiles that reflect GOT2 activity status

Targeting Compensatory Pathways:

  • Low GOT2 expression leads to enhanced nucleotide synthesis and glutathione production , creating potential secondary vulnerabilities.

  • Experimental strategies include:

    • Combined inhibition of glutaminase and nucleotide synthesis pathways

    • Targeting antioxidant systems in GOT2-low tumors

    • Inhibiting transsulfuration pathways that support glutathione synthesis

    • Dual targeting of GOT2-related metabolism and PI3K/AKT/mTOR signaling

Reversing Epigenetic Silencing:

  • For cancers where GOT2 is epigenetically silenced, epigenetic modifiers might restore expression and alter metabolic dependencies .

  • Research approaches include:

    • Screening DNA methyltransferase inhibitors and histone deacetylase inhibitors

    • Testing whether epigenetic restoration of GOT2 sensitizes to conventional therapies

    • Developing combination protocols with sequenced administration of epigenetic and metabolic drugs

Synthetic Lethality Approaches:

  • Identify genes that, when inhibited, cause selective death in GOT2-low cancer cells.

  • Methods include:

    • CRISPR-Cas9 or shRNA library screens in matched GOT2-normal and GOT2-low cell lines

    • Chemical library screens to identify compounds with selective toxicity

    • Computational prediction of synthetic lethal interactions based on metabolic modeling

These approaches highlight how understanding GOT2 biology can reveal targetable metabolic vulnerabilities in cancer, advancing precision medicine strategies for patients with tumors characterized by GOT2 dysregulation .

Product Science Overview

Structure and Function

GOT2 is a homodimeric enzyme, meaning it consists of two identical subunits . The enzyme’s structure includes a large domain that binds pyridoxal phosphate, a small domain, an NH2-terminal arm, and a bridge across two domains . The enzyme catalyzes the transamination of oxaloacetate to aspartate, which is a key step in the malate-aspartate shuttle . This shuttle is essential for transferring reducing equivalents across the mitochondrial membrane, which is crucial for energy production .

Role in Metabolism

GOT2 is involved in several metabolic pathways, including the urea cycle and the tricarboxylic acid cycle . It plays a significant role in maintaining the intracellular NAD(H) redox balance by participating in the malate-aspartate shuttle . This shuttle facilitates the transfer of NADH from the cytosol to the mitochondria, where it can be used in the electron transport chain for ATP production .

Clinical Significance

Mutations in the GOT2 gene have been associated with various diseases, including developmental and epileptic encephalopathy 82 and pheochromocytoma . Additionally, GOT2 has been implicated in cell proliferation and tumor growth . The enzyme’s activity is also linked to the production of kynurenic acid in the brain, which has neuroprotective properties .

Recombinant GOT2

Recombinant human GOT2 is produced using recombinant DNA technology, which involves inserting the human GOT2 gene into a suitable expression system, such as bacteria or yeast . This allows for the production of large quantities of active enzyme for research and therapeutic purposes . Recombinant GOT2 is used in various biochemical assays to study its function and role in different metabolic pathways .

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.