IDH3G Human

Isocitrate Dehydrogenase 3 (NAD+) Gamma Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to IDH3G Human

IDH3G Human refers to the gamma subunit of the mitochondrial NAD⁺-dependent isocitrate dehydrogenase (IDH3), encoded by the IDH3G gene. This enzyme plays a pivotal role in the tricarboxylic acid (TCA) cycle, catalyzing the oxidative decarboxylation of isocitrate to α-ketoglutarate (α-KG), while generating NADH for ATP production . IDH3G is critical for mitochondrial energy metabolism and has emerging roles in redox signaling and cancer biology.

Subunit Composition of IDH3

IDH3 is a heterotetramer comprising:

SubunitGeneMolecular WeightRoleKey Residues
αIDH3A37 kDaCatalytic activity (Arg88, Asp181)Essential for substrate binding and catalysis
βIDH3B39 kDaAllosteric regulation (Arg99, Asp192)Modulates enzyme activity via ADP/NAD⁺ binding
γIDH3G39 kDaRedox-sensitive regulation (Cys148, Cys284)Participates in H₂O₂-mediated metabolic adaptation

Key Functional Features

  • Catalytic Activity: Reversible oxidative decarboxylation of isocitrate to α-KG, utilizing NAD⁺ as a cofactor .

  • Allosteric Regulation: Activated by ADP and inhibited by ATP/NADH, acting as a rate-limiting step in the TCA cycle .

  • Redox Sensitivity: IDH3γ contains critical cysteine residues (Cys148, Cys284) that modulate enzyme activity under oxidative stress, serving as a redox switch .

Redox Signaling and Metabolic Adaptation

IDH3γ functions as a redox-sensitive regulatory node in mitochondrial metabolism:

MechanismEffectExperimental ModelSource
H₂O₂-mediated oxidation of Cys148/284Increased IDH3 activityCardiomyocytes, cancer cell lines (e.g., MDA-MB-231, Hep3B)
Enhanced α-KG productionAlters TCA flux, impacts ATP synthesisMolecular dynamics simulations, cysteine-editing studies

Cancer Biology

ContextRoleEvidenceSource
Glioblastoma (GBM)IDH3α upregulation correlates with tumor progressionIDH3α ablation reduces glycolysis, pentose phosphate shunt usage, and tumorigenicity
Metabolic ReprogrammingIDH3γ interacts with cytosolic SHMT, linking α-KG to nucleotide synthesisIDH3α loss disrupts thymidylate synthesis, elevates SAM levels, and alters DNA methylation
Drug ResistanceIDH3G implicated in gastric cancer drug resistanceAssociation studies (mechanism pending elucidation)

Expression and Localization

IDH3G is ubiquitously expressed in mitochondria across tissues, including:

TissueExpression LevelKey FunctionSource
Heart, Liver, Skeletal MuscleHighTCA cycle activity, ATP production
BrainVariableSynaptic transmission (α-KG modulation of synaptotagmin)

Recombinant IDH3G Production and Applications

ParameterDetailSource
SourceE. coli
Purity>85% (SDS-PAGE)
Formulation20 mM Tris-HCl (pH 8.0), 50% glycerol, 0.2M NaCl
StabilityStore at -20°C; avoid freeze-thaw cycles

Emerging Therapeutic Implications

  1. Neurodegeneration: IDH3α loss in ALS motor neurons disrupts metabolic shifts toward fatty acid oxidation, worsening oxidative stress .

  2. Cancer Metabolism: Targeting IDH3γ redox dynamics may modulate tumor energetics and chemoresistance .

  3. Redox Therapeutics: H₂O₂ signaling via IDH3γ could be leveraged to enhance mitochondrial function in cardiac or metabolic disorders .

Product Specs

Introduction
Isocitrate dehydrogenase [NAD] subunit gamma (IDH3G), found in the mitochondria, belongs to the isocitrate and isopropylmalate dehydrogenases family. Isocitrate dehydrogenases facilitate the oxidative decarboxylation of isocitrate to 2-oxoglutarate. IDH3G is a gamma subunit of an isocitrate dehydrogenase isozyme that specifically uses NAD+ as an electron acceptor and is exclusively located within the mitochondrial matrix.
Description
Recombinant human IDH3G, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 375 amino acids (positions 40-393). It has a molecular weight of 41.1 kDa. A 21 amino acid His-tag is fused to the N-terminus of IDH3G. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The IDH3G solution is provided at a concentration of 0.25 mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0), 50% glycerol, 0.2M NaCl, 5mM DTT, and 2mM EDTA.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is advised for long-term storage. Avoid repeated freezing and thawing of the product.
Purity
Purity is determined to be greater than 85% using SDS-PAGE analysis.
Synonyms
Isocitrate dehydrogenase [NAD] subunit gamma, mitochondrial, Isocitric dehydrogenase subunit gamma, NAD(+)-specific ICDH subunit gamma, IDH3G, H-IDHG.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MFSEQTIPPS AKYGGRHTVT MIPGDGIGPE LMLHVKSVFR HACVPVDFEE VHVSSNADEE DIRNAIMAIR RNRVALKGNI ETNHNLPPSH KSRNNILRTS LDLYANVIHC KSLPGVVTRH KDIDILIVRE NTEGEYSSLE HESVAGVVES LKIITKAKSL RIAEYAFKLA QESGRKKVTA VHKANIMKLG DGLFLQCCRE VAARYPQITF ENMIVDNTTM QLVSRPQQFD VMVMPNLYGN IVNNVCAGLV GGPGLVAGAN YGHVYAVFET ATRNTGKSIA NKNIANPTAT LLASCMMLDH LKLHSYATSI RKAVLASMDN ENMHTPDIGG QGTTSEAIQD VIRHIRVING RAVEA.

Q&A

What is the basic structure and function of IDH3G in human cells?

IDH3G represents the gamma subunit of the IDH3 holoenzyme, which functions within the tricarboxylic acid (TCA) cycle in mitochondria. While the complete crystal structure of the IDH3 holoenzyme remains unreported, biochemical and mutagenesis studies have revealed that the gamma subunit partners with the alpha subunit to form the metal-ICT (isocitrate) binding site . The primary function of IDH3G involves allosteric regulation of the holoenzyme through binding of citrate and ADP . The complete IDH3 enzyme consists of three subunits (α, β, and γ) that form a heterotetramer, with each subunit playing distinct roles: the α subunit binds isocitrate, the β subunit provides structural support through facilitation of subunit assembly, and the γ subunit (IDH3G) regulates enzyme activity .

When properly assembled and functioning, the IDH3 complex catalyzes the oxidative decarboxylation of isocitrate to α-ketoglutarate (αKG) while converting NAD+ to NADH in the process. This reaction represents a critical step in the TCA cycle that generates reducing equivalents (NADH) for the electron transport chain and subsequent ATP production.

How does IDH3G contribute to mitochondrial bioenergetics?

IDH3G plays a crucial role in mitochondrial energy production by regulating the activity of the IDH3 holoenzyme. As the regulatory subunit, IDH3G modulates the rate of isocitrate conversion to α-ketoglutarate and NADH production through allosteric mechanisms . This regulation directly impacts TCA cycle flux and, consequently, the generation of reducing equivalents that feed into oxidative phosphorylation.

Research has demonstrated that IDH3 contributes significantly to cellular respiration and energy production through generation of NADH . In contexts where IDH3 function is compromised, such as in certain neurodegenerative conditions, there is often impaired oxidative phosphorylation and reduced ATP production . Studies in models of amyotrophic lateral sclerosis (ALS) have shown that motor neurons with SOD1 mutations exhibit increased IDH3 expression, suggesting compensatory mechanisms to maintain energy homeostasis when oxidative phosphorylation is impaired.

What methods are commonly used to study IDH3G expression and activity?

Investigation of IDH3G commonly employs a combination of molecular, biochemical, and cellular approaches:

  • Gene Expression Analysis:

    • RT-qPCR to quantify mRNA expression levels

    • RNA sequencing for transcriptomic profiling

    • In situ hybridization for tissue localization

  • Protein Analysis:

    • Western blotting for protein expression quantification

    • Immunohistochemistry/immunofluorescence for tissue and cellular localization

    • Co-immunoprecipitation for protein-protein interaction studies

  • Enzyme Activity Assays:

    • Spectrophotometric assays measuring NAD+ to NADH conversion

    • Metabolite analysis using mass spectrometry to track isocitrate conversion to α-ketoglutarate

    • Oxygen consumption rate measurements to assess impact on mitochondrial respiration

  • Genetic Manipulation:

    • RNA interference or CRISPR-Cas9 techniques for knockdown/knockout studies

    • Overexpression systems using plasmid transfection

    • Site-directed mutagenesis to study specific residues involved in regulation

The choice of method depends on the specific research question, with experimental designs often combining multiple approaches to provide comprehensive insights into IDH3G function and regulation.

How does IDH3G differ from other IDH isoforms?

IDH3G differs substantially from other IDH isoforms in structure, cofactor preference, subcellular localization, and function:

FeatureIDH3G (as part of IDH3)IDH1IDH2
Subcellular locationMitochondriaCytosol/peroxisomesMitochondria
CofactorNAD+NADP+NADP+
Reaction reversibilityIrreversibleReversibleReversible
Subunit compositionPart of heterotetramer (α₂βγ)HomodimerHomodimer
Role in regulationAllosteric regulationNo equivalent regulatory subunitNo equivalent regulatory subunit
Common mutations in cancerRareR132H common in gliomasR172K in AML

IDH3, containing the IDH3G subunit, exclusively uses NAD+ as a cofactor and catalyzes an irreversible reaction in the TCA cycle, while IDH1 and IDH2 utilize NADP+ and catalyze reversible reactions . The most significant distinction is that IDH3G serves as a regulatory subunit within a heteromeric complex, whereas IDH1 and IDH2 form homomeric enzymes . While cancer-associated mutations are well-documented in IDH1 and IDH2, leading to production of the oncometabolite (R)-2-hydroxyglutarate, mutations in IDH3G and other IDH3 subunits are less frequently observed in cancer.

What are the current experimental approaches to investigate IDH3G's role in disease pathologies?

Advanced investigation of IDH3G's role in disease pathologies employs sophisticated methodological approaches:

  • Patient-Derived Models:

    • Primary cell cultures from patient biopsies

    • Patient-derived xenografts in immunocompromised mice

    • Induced pluripotent stem cells (iPSCs) differentiated into relevant cell types

  • Advanced Genetic Engineering:

    • Conditional knock-in/knockout models with tissue-specific or temporal control

    • CRISPR-Cas9 base editing for precise introduction of patient-specific mutations

    • AAV-mediated gene delivery for in vivo studies

  • Multi-omics Integration:

    • Metabolomics to profile TCA cycle intermediates and related metabolites

    • Proteomics to identify post-translational modifications and interaction partners

    • Transcriptomics to identify downstream gene expression changes

    • Integration of datasets using computational biology approaches

  • Functional Metabolic Analysis:

    • Isotope tracing with 13C-labeled substrates to track metabolic flux

    • Seahorse XF analysis for real-time measurements of oxygen consumption and extracellular acidification

    • In vivo metabolic imaging using positron emission tomography

  • Structural Biology Approaches:

    • Cryo-electron microscopy to resolve the structure of the IDH3 complex

    • Hydrogen-deuterium exchange mass spectrometry to study conformational dynamics

    • Molecular dynamics simulations to predict functional consequences of mutations

These approaches are often combined to comprehensively understand how alterations in IDH3G contribute to disease mechanisms and to identify potential therapeutic interventions targeting this enzyme or its regulatory pathways.

How do post-translational modifications regulate IDH3G function?

Post-translational modifications (PTMs) of IDH3G represent a sophisticated regulatory layer that fine-tunes enzyme activity in response to metabolic demands and cellular stress. While the search results don't explicitly detail IDH3G-specific PTMs, research on the IDH3 complex suggests several regulatory mechanisms:

Experimental approaches to study these modifications include targeted mass spectrometry, use of phospho-specific antibodies, and pharmacological manipulation of modifying enzymes coupled with functional assays of IDH3 activity. Understanding the PTM landscape of IDH3G is crucial for developing comprehensive models of TCA cycle regulation in both normal physiology and disease states.

What cell-type specific functions of IDH3G have been identified in neuronal cells?

IDH3G exhibits several cell-type specific functions in neuronal cells that highlight its importance in brain metabolism and neurodegeneration:

  • Synaptic Transmission Regulation:

    • Studies in Drosophila have shown that IDH3α (which partners with IDH3G) is critical for normal synaptic transmission

    • Loss of IDH3α function impairs synaptic transmission in photoreceptors and larval neuromuscular junctions, which can be restored by α-ketoglutarate supplementation

    • α-Ketoglutarate enhances synaptotagmin1 (Syt1)-lipid interaction and increases vesicle fusion, processes that would be compromised when IDH3 function is impaired

  • Protection Against Oxidative Stress:

    • Neurons are particularly vulnerable to oxidative damage due to high metabolic rates and limited regenerative capacity

    • IDH3, as part of the TCA cycle, contributes to maintaining NADH levels which indirectly support glutathione regeneration systems

    • In models of Parkinson's disease, IDH function has been linked to protection against ROS accumulation, with both IDH1 and IDH2 showing cytoprotective effects in dopaminergic neurons

  • Metabolic Adaptation in Neurodegeneration:

    • In amyotrophic lateral sclerosis (ALS) models with SOD1 mutations, IDH3 expression is upregulated as motor neurons shift toward increased TCA cycle activity to compensate for impaired oxidative phosphorylation

    • This metabolic shift facilitates increased utilization of fatty acids for energy production

  • Developmental Neuronal Function:

    • Homozygous mutations in IDH3A (which partners with IDH3G) have been associated with severe epileptic encephalopathy in infants, suggesting critical roles for the IDH3 complex in neuronal development and function

These findings underscore the multifaceted roles of the IDH3 complex, including the IDH3G subunit, in neuronal function beyond basic metabolic support, with implications for understanding and potentially treating neurodegenerative conditions.

How does IDH3G contribute to metabolic reprogramming in cancer cells?

While IDH1 and IDH2 mutations are well-established cancer drivers, the role of IDH3G in metabolic reprogramming of cancer cells is more nuanced and less directly studied. Based on research on the IDH3 complex:

  • Altered Expression Patterns:

    • IDH3α (which functions with IDH3G) is upregulated in several cancers including glioblastoma, particularly at the tumor's leading edge and in tumor-associated endothelium

    • Changes in IDH3G expression could potentially influence the assembly and function of the IDH3 holoenzyme

  • TCA Cycle Flux Modulation:

    • Ablation of IDH3α reduces carbon flux through the TCA cycle, inducing a metabolic shift toward increased glycolysis and pentose phosphate pathway utilization

    • This metabolic rewiring reduces the tumorigenic potential of transformed astrocytes and patient-derived glioma-initiating cells

    • As part of the IDH3 complex, IDH3G likely contributes to this regulation of TCA cycle activity

  • Extramitochondrial Functions:

    • IDH3α shows cell cycle-dependent extramitochondrial localization, with predominant accumulation in the cytosol and nuclear periphery during S phase

    • In these locations, IDH3α interacts with cytosolic serine hydroxymethyltransferase (cSHMT), influencing the de novo thymidylate synthesis pathway

    • IDH3G may also exhibit non-canonical functions outside mitochondria, potentially contributing to cancer cell adaptation

  • Epigenetic Influence:

    • Loss of IDH3α expression affects cSHMT function, reducing thymidylate synthesis rates while increasing carbon flux into the methionine salvage pathway

    • This leads to elevated levels of the methyl donor S-adenosyl methionine and increased DNA methylation

    • Through its regulatory effect on the IDH3 holoenzyme, IDH3G could indirectly influence these epigenetic mechanisms

  • Compartmentalized TCA Cycle Function:

    • Multiple TCA enzymes, including components of IDH3, can localize to the cell nucleus while maintaining mitochondrial pools during both embryonic development and tumorigenesis

    • This differential compartmentalization allows fine-tuning of TCA cycle activity to regulate energy production, macromolecular synthesis, and gene expression in response to environmental and developmental cues

Understanding how IDH3G contributes to these processes could potentially reveal new therapeutic targets for cancers where metabolic reprogramming is a key driver of tumor progression.

What experimental challenges exist in studying IDH3G-specific functions?

Researchers face several significant challenges when investigating IDH3G-specific functions:

To overcome these challenges, researchers should consider combinatorial approaches:

  • Employing inducible and tissue-specific genetic models

  • Utilizing CRISPR-based approaches for precise manipulation of IDH3G

  • Applying multi-omics strategies that integrate metabolomics, proteomics, and transcriptomics data

  • Developing computational models that account for pathway interactions and compensatory mechanisms

  • Establishing in vitro reconstitution systems with purified components to dissect specific biochemical functions

What are the most effective systems for modeling IDH3G dysfunction in neurological disorders?

Modeling IDH3G dysfunction in neurological disorders requires carefully selected experimental systems that recapitulate relevant aspects of human pathophysiology:

  • Cellular Models:

    • iPSC-Derived Neurons: Patient-derived induced pluripotent stem cells differentiated into relevant neuronal subtypes provide physiologically relevant models with genetic backgrounds matching affected individuals

    • Primary Neuronal Cultures: Cultures from rodent models with genetic modifications in IDH3G allow detailed investigation of cellular phenotypes

    • Neuronal Cell Lines with CRISPR-Edited IDH3G: Engineered neuroblastoma or other neuronal cell lines with precise modifications to IDH3G

  • Invertebrate Models:

    • Drosophila Models: Studies have already established that IDH dysfunction in Drosophila results in neurodegeneration resembling PD, with increased ROS and locomotive dysfunction

    • C. elegans: Transparent nematodes allow for visualization of neurons while manipulating IDH3G orthologs

  • Vertebrate Models:

    • Conditional Knockout Mice: Tissue-specific and inducible deletion of IDH3G in relevant neuronal populations

    • Transgenic Models: Expression of mutant IDH3G variants identified in human patients

    • AAV-Mediated Gene Delivery: Viral delivery of IDH3G mutants or shRNAs to specific brain regions

  • Ex Vivo Systems:

    • Organotypic Brain Slice Cultures: Preserve tissue architecture while allowing experimental manipulation

    • Brain Organoids: 3D cultures that recapitulate aspects of brain development and neuronal connectivity

  • Computational Models:

    • Metabolic Flux Analysis Models: Computational simulations of altered TCA cycle dynamics

    • Neural Network Models: Predictions of how metabolic alterations affect neuronal circuit function

Each model system offers distinct advantages:

Model SystemAdvantagesLimitationsBest Applications
iPSC-derived neuronsHuman genetic background, disease-relevant mutationsVariable differentiation, lack tissue contextPatient-specific drug screening, molecular mechanisms
DrosophilaRapid generation time, powerful geneticsEvolutionary distance from humansInitial functional screening, in vivo phenotypes
Conditional mouse modelsMammalian physiology, brain complexityTime and resource intensiveIn vivo disease progression, complex behaviors
Brain organoids3D organization, multiple cell typesLack vascularization, variabilityDevelopmental effects, cell-cell interactions

The optimal approach often involves complementary use of multiple model systems, with simpler models for initial mechanistic studies and more complex systems to validate findings in contexts more closely resembling human pathophysiology.

How can researchers effectively measure IDH3G-specific metabolic flux in living cells?

Measuring IDH3G-specific metabolic flux in living cells presents considerable technical challenges but can be accomplished through sophisticated methodological approaches:

  • Isotope Tracing with Mass Spectrometry:

    • 13C-labeled substrates: Using labeled glucose, glutamine, or other TCA cycle intermediates to track carbon flow specifically through IDH3-catalyzed reactions

    • Positional isotopomer analysis: Examining the pattern of labeled carbon atoms in α-ketoglutarate to distinguish IDH3-specific activity from other IDH isoforms

    • Time-course sampling: Capturing dynamic changes in metabolite labeling patterns to infer flux rates

  • Genetic and Pharmacological Approaches:

    • Isoform-specific knockdown/knockout: Using targeted siRNA or CRISPR to specifically reduce IDH3G expression while monitoring metabolic consequences

    • Combined inhibition strategies: Selective inhibition of IDH1 and IDH2 to isolate IDH3-dependent metabolism

    • Rescue experiments: Reintroduction of wild-type or mutant IDH3G to determine functional consequences

  • Real-time Metabolic Measurements:

    • NAD+/NADH ratio sensors: Fluorescent protein-based sensors to monitor changes in NAD+/NADH ratios in mitochondria

    • Oxygen consumption rate (OCR): Measuring respiratory changes in response to IDH3G manipulation

    • Mitochondrial membrane potential: Tracking changes in mitochondrial energetics

  • Subcellular Resolution Techniques:

    • Metabolite imaging: Using chemical probes or genetically encoded sensors for spatiotemporal resolution of metabolite concentrations

    • Single-cell metabolomics: Analyzing metabolic profiles at the individual cell level to account for cellular heterogeneity

    • Organelle-specific isolation: Purifying mitochondria before metabolic analysis to enrich for IDH3-specific activity

  • Computational Integration:

    • Flux balance analysis (FBA): Mathematical modeling to predict metabolic flux distribution

    • Kinetic modeling: Incorporating enzyme kinetic parameters to simulate IDH3G activity

    • Multi-omics data integration: Combining metabolomics with proteomics and transcriptomics to create comprehensive models

Experimental Protocol Example:

  • Generate cell lines with doxycycline-inducible IDH3G knockdown

  • Pre-treat cells with selective IDH1/2 inhibitors to minimize compensatory flux

  • Introduce 13C5-glutamine as a metabolic tracer

  • Collect samples at multiple time points (5, 15, 30, 60, 120 minutes)

  • Analyze isotopologue distribution using LC-MS/MS

  • Apply computational modeling to determine flux rates

  • Validate with orthogonal approaches such as oxygen consumption measurements

This integrated approach allows researchers to distinguish IDH3G-specific contributions to TCA cycle flux from those of other IDH isoforms and to evaluate the impact of genetic or pharmacological interventions on IDH3G function in living cells.

What are the best approaches for studying IDH3G protein-protein interactions and complex assembly?

Investigating IDH3G protein-protein interactions and complex assembly requires a combination of biochemical, biophysical, and cellular approaches:

  • Affinity Purification Coupled with Mass Spectrometry:

    • Tandem Affinity Purification (TAP): Using dual tags on IDH3G to achieve high purity of protein complexes

    • BioID or APEX2 Proximity Labeling: Identifying proteins in close proximity to IDH3G through biotinylation

    • Cross-linking Mass Spectrometry (XL-MS): Capturing transient interactions through chemical cross-linking

  • Advanced Microscopy Techniques:

    • Förster Resonance Energy Transfer (FRET): Measuring protein-protein interactions in living cells

    • Fluorescence Correlation Spectroscopy (FCS): Analyzing diffusion properties to infer complex formation

    • Stimulated Emission Depletion (STED) Microscopy: Super-resolution imaging of IDH3G complexes

  • Protein Complementation Assays:

    • Split Luciferase Complementation: Reconstitution of luciferase activity when two tagged proteins interact

    • Bimolecular Fluorescence Complementation (BiFC): Fluorescent signal generation upon protein interaction

    • Mammalian Two-Hybrid System: Detecting interactions through transcriptional reporter activation

  • Biochemical and Biophysical Methods:

    • Blue Native PAGE: Preserving native protein complexes during electrophoretic separation

    • Size Exclusion Chromatography with Multi-Angle Light Scattering (SEC-MALS): Determining complex stoichiometry and molecular weight

    • Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS): Mapping interaction interfaces and conformational changes

    • Surface Plasmon Resonance (SPR) or Bio-Layer Interferometry (BLI): Measuring binding kinetics and affinities

  • Structural Biology Approaches:

    • Cryo-Electron Microscopy: Resolving structures of IDH3 complexes, particularly important since "the crystal structure of the IDH3 holoenzyme has not been reported yet"

    • Small-Angle X-ray Scattering (SAXS): Obtaining low-resolution structural information in solution

    • Integrative Structural Modeling: Combining multiple data types to generate structural models

Experimental Strategy Example:

To systematically study IDH3G assembly into the IDH3 holoenzyme:

  • Generate constructs expressing IDH3G with various affinity tags (e.g., FLAG, HA, BioID)

  • Perform staged purifications to capture assembly intermediates

  • Use cross-linking to stabilize transient interactions

  • Analyze by mass spectrometry to identify interaction partners and post-translational modifications

  • Validate key interactions using microscopy-based methods in living cells

  • Map interaction domains through mutagenesis studies

  • Determine structural features using cryo-EM or integrative modeling approaches

This multi-faceted approach would provide comprehensive insights into how IDH3G contributes to the assembly and regulation of the IDH3 holoenzyme, potentially revealing novel regulatory mechanisms and therapeutic targets.

How can researchers differentiate the specific contributions of IDH3G from other IDH isoforms in metabolic studies?

Distinguishing IDH3G-specific functions from other IDH isoforms requires carefully designed experimental strategies that exploit unique features of each enzyme:

  • Cofactor-Specific Approaches:

    • NAD+ vs. NADP+ Discrimination: IDH3 exclusively uses NAD+ while IDH1 and IDH2 use NADP+

    • Cofactor Manipulation: Altering cellular NAD+/NADP+ ratios to preferentially affect specific isoforms

    • Activity Assays: Designing reaction conditions that selectively measure NAD+-dependent (IDH3) or NADP+-dependent (IDH1/2) activity

  • Genetic Manipulation Strategies:

    • Isoform-Specific Knockdown/Knockout: Using siRNA, shRNA, or CRISPR-Cas9 targeting unique regions of IDH3G

    • Rescue Experiments: Complementation with wild-type or mutant IDH3G in knockout backgrounds

    • Inducible Systems: Temporal control of IDH3G expression to observe acute metabolic changes

  • Subcellular Fractionation and Localization:

    • Mitochondrial Isolation: Enriching for IDH3-containing compartments

    • Compartment-Specific Sensors: Using targeted metabolite sensors to monitor activity in specific cellular locations

    • Immunofluorescence with Isoform-Specific Antibodies: Visualizing differential localization patterns

  • Metabolic Flux Analysis:

    • Reaction Directionality: IDH3 catalyzes an irreversible reaction, while IDH1/2 reactions are reversible

    • Isotopomer Pattern Analysis: Examining labeling patterns that distinguish forward vs. reverse flux

    • Mathematical Modeling: Computational approaches to deconvolute isoform-specific contributions to measured fluxes

  • Pharmacological Approaches:

    • Isoform-Selective Inhibitors: Using compounds that specifically target IDH1/2 to isolate IDH3 function

    • Allosteric Modulators: Targeting the unique regulatory properties of IDH3G

    • Combination Strategies: Sequential inhibition to determine hierarchy of metabolic compensation

Experimental Design Example:

To distinguish IDH3G-specific metabolic effects in cells:

  • Preparation Phase:

    • Generate cell lines with doxycycline-inducible IDH3G knockdown

    • Create matched control lines with IDH1 or IDH2 knockdown

    • Validate isoform-specific targeting using qPCR and western blotting

  • Experimental Phase:

    • Culture cells with 13C-labeled glucose or glutamine

    • Induce knockdown of specific IDH isoforms

    • Collect samples for metabolomic analysis at multiple time points

    • Perform parallel measurements of oxygen consumption and extracellular acidification

  • Analysis Phase:

    • Quantify isotopomer distributions in TCA cycle intermediates

    • Compare metabolic profiles between IDH3G-depleted and control cells

    • Apply computational modeling to determine isoform-specific flux contributions

    • Validate findings with orthogonal approaches such as enzyme activity assays

This comprehensive approach leverages the unique characteristics of IDH3G—its NAD+ dependence, irreversible catalytic activity, mitochondrial localization, and heterotetrameric structure—to distinguish its specific contributions from those of other IDH isoforms in cellular metabolism.

How do alterations in IDH3G expression contribute to neurodegenerative disease progression?

Alterations in IDH3G expression and function have emerging roles in neurodegenerative disease pathogenesis through several interconnected mechanisms:

  • Redox Homeostasis Disruption:

    • IDH enzymes are critical for maintaining cellular redox balance through generation of NADPH (IDH1/2) and NADH (IDH3)

    • Reduced IDH function compromises glutathione regeneration systems, leading to increased oxidative damage to lipids, proteins, and nucleic acids—a hallmark of Parkinson's disease, Alzheimer's disease, and other neurodegenerative conditions

    • In Drosophila models, IDH deficiency phenocopies DJ-1 loss-of-function, showing increased ROS, dopaminergic neuron degeneration, and impaired locomotive function

  • Mitochondrial Dysfunction:

    • As a component of the IDH3 complex, IDH3G plays a central role in TCA cycle function and mitochondrial energy production

    • Altered IDH3 expression affects oxidative phosphorylation efficiency, which is particularly detrimental to neurons with high energy demands

    • Impaired mitochondrial function leads to energy deficits that compromise neuronal survival and function

  • Metabolic Rewiring in Neurodegeneration:

    • In ALS motor neurons with SOD1 mutations, IDH3 expression is upregulated as part of a compensatory metabolic shift toward increased TCA cycle activity and fatty acid oxidation

    • This metabolic adaptation helps neurons meet energy requirements when oxidative phosphorylation is compromised

    • Failure of these compensatory mechanisms may contribute to disease progression

  • Synaptic Dysfunction:

    • IDH3 function and α-ketoglutarate production are linked to synaptic transmission through effects on synaptotagmin1 dynamics

    • α-Ketoglutarate enhances Syt1-lipid interaction and SV fusion with the plasma membrane

    • Reduced IDH3 function may therefore directly impair neurotransmission, contributing to cognitive and motor symptoms

  • Epigenetic Dysregulation:

    • IDH3-generated α-ketoglutarate serves as a cofactor for numerous epigenetic modifying enzymes

    • Altered α-ketoglutarate levels affect histone and DNA demethylation processes, potentially disrupting neuronal gene expression patterns

    • These epigenetic changes may contribute to altered neuronal identity and function in neurodegenerative conditions

Monitoring disease progression in relation to IDH3G status requires longitudinal studies combining clinical assessments with biomarker measurements (such as α-ketoglutarate levels, redox status indicators, and metabolic profiles) and neuroimaging to track disease-related changes in brain structure and function.

What are the challenges in targeting IDH3G therapeutically for metabolic disorders?

Developing therapeutic strategies targeting IDH3G presents several significant challenges that researchers must address:

  • Target Specificity and Selectivity:

    • Isoform Selectivity: Designing compounds that selectively modulate IDH3G without affecting IDH1/2

    • Subunit Specificity: Targeting the gamma subunit specifically within the IDH3 complex

    • Allosteric Modulation: Identifying druggable allosteric sites unique to IDH3G

  • Physiological Redundancy and Compensation:

    • Metabolic Plasticity: Cells can rewire metabolism through alternative pathways when IDH3 is inhibited

    • Isoform Redundancy: IDH1/2 may partially compensate for altered IDH3 function

    • Tissue-Specific Effects: IDH3G modulation may have distinct consequences in different tissues

  • Therapeutic Window Considerations:

    • Essential Metabolic Function: Complete inhibition of IDH3G may be poorly tolerated due to its central role in energy metabolism

    • Tissue-Specific Requirements: Neurons and other high-energy tissues may be particularly sensitive to IDH3 modulation

    • Disease Context Dependency: Optimal degree of modulation likely varies based on specific disorder

  • Delivery and Pharmacokinetic Challenges:

    • Mitochondrial Targeting: Delivering compounds to mitochondria where IDH3G functions

    • Blood-Brain Barrier: Achieving sufficient CNS penetration for neurological indications

    • Sustained Modulation: Maintaining therapeutic levels to achieve lasting metabolic effects

  • Clinical Development Considerations:

    • Biomarker Development: Identifying reliable indicators of IDH3G modulation (e.g., α-ketoglutarate/isocitrate ratios)

    • Patient Stratification: Determining which patients are most likely to benefit from IDH3G-targeted therapy

    • Combination Approaches: Potentially combining IDH3G modulation with other metabolic interventions

Unlike IDH1 and IDH2 mutations in cancer, which produce the oncometabolite (R)-2-hydroxyglutarate and have clear therapeutic rationales with FDA-approved inhibitors, targeting IDH3G may require more nuanced approaches focusing on modulation rather than inhibition. Potential therapeutic strategies could include:

  • Allosteric Activators: Enhancing IDH3 function in conditions with reduced TCA cycle activity

  • Metabolic Bypass Strategies: Supplementing α-ketoglutarate to compensate for IDH3 dysfunction

  • Gene Therapy: Correcting genetic defects in IDH3G in hereditary disorders

  • RNA-Based Approaches: Using antisense oligonucleotides or siRNA for precise modulation of expression levels

The optimal therapeutic approach will depend on whether the pathology involves insufficient or excessive IDH3G activity, the specific tissues affected, and the underlying disease mechanism.

How can IDH3G be leveraged as a biomarker in metabolic and neurological disorders?

IDH3G and related metabolic parameters have significant potential as biomarkers for various disorders:

  • Direct Measurement Approaches:

    • Protein Expression: Quantifying IDH3G levels in accessible tissues or biofluids

    • Genetic Analysis: Screening for mutations or polymorphisms in IDH3G

    • Post-translational Modifications: Assessing phosphorylation, acetylation, or other modifications that affect IDH3G activity

  • Metabolic Signatures:

    • α-Ketoglutarate/Isocitrate Ratio: Directly reflects IDH3 activity

    • NAD+/NADH Balance: Indicates TCA cycle function and redox state

    • Comprehensive Metabolomic Profiling: Identifying broader metabolic patterns associated with IDH3G dysfunction

  • Disease-Specific Applications:

    • Neurodegenerative Disorders:

      • Cerebrospinal fluid metabolite profiling

      • PET imaging of brain metabolism

      • Correlation with cognitive or motor function measures

    • Metabolic Disorders:

      • Serum/plasma metabolite analysis

      • Exercise challenge tests to assess TCA cycle reserve

      • Integration with other metabolic parameters (glucose, lactate, pyruvate)

  • Technical Considerations:

    • Sample Collection: Standardizing conditions to minimize preanalytical variability

    • Analytical Methods: Employing sensitive mass spectrometry or NMR-based approaches

    • Reference Ranges: Establishing age, sex, and population-specific normal values

  • Clinical Implementation Strategies:

    • Risk Stratification: Identifying patients at higher risk for disease progression

    • Treatment Response Monitoring: Tracking metabolic changes during therapeutic interventions

    • Combination Biomarker Panels: Integrating IDH3G-related markers with other disease indicators

While IDH1 mutations in cancer can be monitored through (R)-2-hydroxyglutarate measurements in serum with prognostic value (levels >200 ng/ml correlate with poorer outcomes), IDH3G-related biomarkers would likely focus on TCA cycle intermediate levels and ratios. The approach would differ based on the disease context:

Disease ContextPotential BiomarkersSample TypesClinical Applications
Neurodegenerative diseasesα-KG/isocitrate ratio, NAD+/NADH, glutathione statusCSF, plasma, PET imagingEarly detection, progression monitoring
Metabolic disordersTCA cycle intermediates, energy charge (ATP/ADP/AMP)Plasma, urine, tissue biopsiesDiagnosis, treatment response
Mitochondrial diseasesRespiratory chain activity, IDH3 complex assemblyMuscle biopsies, fibroblastsClassification, severity assessment

Longitudinal biomarker monitoring combined with clinical assessments would provide the most valuable information for patient management and therapeutic decision-making.

What translational research approaches show promise for IDH3G-related diseases?

Several promising translational research approaches are emerging for IDH3G-related diseases:

For neurodegenerative conditions where IDH3 dysfunction contributes to pathology, promising approaches include:

  • Targeting Redox Imbalance: Since IDH dysfunction is linked to impaired redox homeostasis in PD, AD, and other conditions, therapies that restore glutathione levels or provide alternative antioxidant support may be beneficial

  • Enhancing Metabolic Flexibility: In ALS models where IDH3 is upregulated to facilitate metabolic adaptation, therapies that further enhance this compensatory shift or provide alternative energy substrates could potentially slow disease progression

  • Supporting Synaptic Function: Given the role of α-ketoglutarate in enhancing synaptotagmin1-mediated vesicle fusion, targeted delivery of α-ketoglutarate or compounds that enhance local production could potentially improve synaptic transmission in relevant disorders

These translational approaches should be evaluated in appropriate model systems before advancing to clinical trials, with careful consideration of disease-specific mechanisms and potential tissue-specific effects of IDH3G modulation.

What emerging technologies will advance our understanding of IDH3G regulation?

Several cutting-edge technologies are poised to revolutionize our understanding of IDH3G regulation and function:

  • Advanced Single-Cell Technologies:

    • Single-Cell Metabolomics: Capturing cell-to-cell variability in metabolite levels and IDH3-related metabolic activities

    • Spatial Metabolomics: Mapping metabolic profiles with spatial resolution in tissues

    • Multi-modal Single-Cell Analysis: Simultaneously measuring transcriptome, proteome, and metabolome in the same cells

  • Real-Time Dynamic Monitoring Systems:

    • Genetically Encoded Metabolite Sensors: Fluorescent reporters specifically designed to monitor TCA cycle intermediates

    • NAD+/NADH Ratio Biosensors: Real-time tracking of redox status in living cells

    • Activity-Based Protein Sensors: Direct measurement of IDH3G conformational changes and activity states

  • Structural Biology Innovations:

    • Cryo-Electron Tomography: Visualizing IDH3 complexes in their native cellular environment

    • 4D Structural Biology: Capturing dynamic structural changes during enzyme catalysis

    • Integrative Structural Approaches: Combining multiple data types to model complete IDH3 holoenzyme structure, which "has not been reported yet"

  • Genome Engineering Advances:

    • Base Editing and Prime Editing: Creating precise mutations to study structure-function relationships

    • Epigenome Editing: Manipulating IDH3G expression through targeted epigenetic modifications

    • Spatial and Temporal Control: Optogenetic or chemogenetic regulation of IDH3G activity

  • AI and Computational Approaches:

    • Deep Learning for Metabolic Modeling: Predicting metabolic responses to IDH3G perturbations

    • Network Analysis: Identifying regulatory relationships between IDH3G and other cellular components

    • Multi-Scale Modeling: Integrating molecular, cellular, and tissue-level effects of IDH3G function

These technologies will help address key outstanding questions:

  • How do post-translational modifications dynamically regulate IDH3G in response to changing metabolic demands?

  • What are the structural determinants of IDH3G's allosteric regulation through citrate and ADP binding?

  • How does IDH3G contribute to non-canonical functions of the IDH3 complex in different subcellular compartments?

  • What cell-type specific regulatory mechanisms control IDH3G expression and function in diverse tissues?

The integration of these technologies will enable a systems biology view of IDH3G regulation, potentially revealing novel therapeutic approaches for disorders involving mitochondrial dysfunction, altered TCA cycle activity, or disrupted cellular redox balance.

How might artificial intelligence accelerate IDH3G-focused drug discovery?

Artificial intelligence (AI) offers transformative potential for accelerating IDH3G-targeted therapeutic development through multiple complementary approaches:

  • Structure-Based Drug Design:

    • Protein Structure Prediction: Tools like AlphaFold2 can generate high-confidence models of IDH3G structure and the complete IDH3 holoenzyme, addressing the gap that "the crystal structure of the IDH3 holoenzyme has not been reported yet"

    • Binding Site Identification: ML algorithms can identify novel druggable pockets, particularly allosteric sites related to IDH3G's regulatory function

    • Virtual Screening: Deep learning models can rapidly screen millions of compounds to identify potential IDH3G modulators

    • De Novo Molecule Generation: Generative models can design entirely new chemical entities optimized for IDH3G binding

  • Target Validation and Disease Modeling:

    • Multi-omics Data Integration: AI can identify disease signatures associated with IDH3G dysfunction across genomic, transcriptomic, proteomic, and metabolomic datasets

    • Patient Stratification: Unsupervised learning approaches can identify patient subgroups most likely to benefit from IDH3G-targeted therapies

    • Disease Progression Modeling: Predicting the impact of IDH3G modulation on disease trajectories

  • Preclinical and Clinical Development:

    • ADMET Prediction: ML models can forecast absorption, distribution, metabolism, excretion, and toxicity properties of candidate compounds

    • Drug Repurposing: Identifying existing approved drugs that may modulate IDH3G function

    • Combination Therapy Optimization: Predicting synergistic drug combinations that include IDH3G modulators

    • Clinical Trial Design: Optimizing patient selection and endpoint measurements for IDH3G-targeted interventions

  • Mechanistic Insights:

    • Metabolic Network Modeling: Predicting system-wide effects of IDH3G modulation

    • Protein-Protein Interaction Networks: Identifying novel interaction partners and regulatory relationships

    • Dynamic Simulation: Modeling the temporal aspects of IDH3G regulation in response to metabolic perturbations

AI-driven workflow examples for IDH3G drug discovery:

  • Target Engagement Focus:

    • Generate predicted structures of IDH3G alone and within the holoenzyme complex

    • Identify allosteric pockets unique to the gamma subunit

    • Design small molecules that specifically modulate IDH3G's regulatory function

    • Optimize candidates for blood-brain barrier penetration for neurological indications

  • Phenotypic Screening Approach:

    • Train deep learning models on cellular responses to thousands of compounds

    • Identify patterns associated with normalized TCA cycle function

    • Screen compound libraries for molecules producing desired metabolic signatures

    • Validate hits through targeted assays of IDH3 function

  • Patient-Specific Therapy Design:

    • Analyze patient-derived cells using multi-omics approaches

    • Build AI models relating metabolic profiles to disease phenotypes

    • Predict patient-specific responses to IDH3G modulators

    • Design personalized combination therapies based on individual metabolic signatures

These AI-driven approaches could dramatically reduce the time and resources required to develop effective IDH3G-targeted therapeutics while increasing the probability of clinical success through more precise understanding of target biology and patient selection.

Product Science Overview

Structure and Function

IDH3G is a subunit of the isocitrate dehydrogenase enzyme complex, which catalyzes the oxidative decarboxylation of isocitrate to 2-oxoglutarate (alpha-ketoglutarate) while reducing NAD+ to NADH . This reaction is a critical step in the citric acid cycle, contributing to cellular respiration and energy production.

The human recombinant form of IDH3G is produced in Escherichia coli (E. coli) and is a single, non-glycosylated polypeptide chain containing 375 amino acids. It has a molecular mass of approximately 41.1 kDa and is fused to a 21 amino acid His-tag at the N-terminus . This recombinant protein is purified using proprietary chromatographic techniques to ensure high purity and functionality .

Biological Significance

IDH3G is essential for the proper functioning of the citric acid cycle. The enzyme’s activity is tightly regulated to maintain metabolic balance within the cell. Mutations or dysregulation of IDH3G can lead to metabolic disorders and have been associated with various diseases, including D-2-Hydroxyglutaric Aciduria 2 and Pseudopseudohypoparathyroidism .

Applications

The recombinant form of IDH3G is widely used in research to study its structure, function, and role in metabolism. It is also utilized in drug discovery and development, particularly in the context of metabolic diseases and cancer research. The availability of high-purity recombinant IDH3G allows scientists to conduct detailed biochemical and biophysical analyses, facilitating a deeper understanding of its mechanisms and interactions.

Storage and Stability

The recombinant IDH3G protein is typically stored at -20°C for long-term preservation. It is recommended to add a carrier protein, such as human serum albumin (HSA) or bovine serum albumin (BSA), to prevent degradation during storage. The protein should be handled with care to avoid multiple freeze-thaw cycles, which can affect its stability and activity .

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.