Isocitrate dehydrogenase 1 (IDH1) is a cytoplasmic enzyme critical for cellular metabolism, redox balance, and epigenetic regulation. In humans, it catalyzes the oxidative decarboxylation of isocitrate to α-ketoglutarate (α-KG), generating NADPH as a byproduct. Beyond its metabolic role, IDH1 mutations are strongly associated with myeloid malignancies and gliomas, where they drive oncogenesis through oncometabolite production and epigenetic dysregulation.
IDH1 exists as a homodimer, with each subunit containing a Rossmann fold (large domain), α/β sandwich (small domain), and β-sheet clasp domain . It localizes to the cytoplasm, peroxisomes, and endoplasmic reticulum, where it:
Converts isocitrate → α-KG + NADPH: Central to the TCA cycle and antioxidant defense.
Maintains redox homeostasis: NADPH production mitigates oxidative stress.
Supports β-oxidation: Critical for unsaturated fatty acid metabolism in the liver .
Enzymatic Activity | Wild-Type IDH1 | Mutant IDH1 |
---|---|---|
Substrate | Isocitrate | α-KG |
Product | α-KG + NADPH | D-2-HG + NADP⁺ |
Role | Metabolism/redox | Oncogenesis |
IDH1 mutations (e.g., R132H) are neomorphic, causing the enzyme to produce the oncometabolite D-2-hydroxyglutarate (D-2-HG). This disrupts α-KG-dependent dioxygenases, leading to DNA hypermethylation and impaired differentiation .
IDH1 Status | Median Survival (Months) | Progression-Free Survival (Months) |
---|---|---|
Mutated | 207 | 100 |
Wild-type | 25 | 9 |
Data from TCGA glioma cohort (n=921) . |
Recent studies highlight IDH1’s non-enzymatic roles:
IDH1 knockdown in human erythroblasts causes:
Aberrant nuclear condensation: 2-fold increase in nucleus-to-plasma ratio .
Chromatin state changes: Genome-wide redistribution of H3K79me3, linked to SIRT1 downregulation .
Impaired enucleation: Accumulation of polychromatic/orthochromatic erythroblasts .
Category | Upregulated Genes | Downregulated Genes |
---|---|---|
Chromatin remodeling | 3,543 (e.g., H3K79me3 regulators) | – |
Metabolism | – | 3,295 (e.g., SIRT1) |
RNA-seq data from erythroid cells . |
α-KG supplementation: Fails to rescue defects in erythropoiesis, indicating non-enzymatic roles .
ROS scavenging: No improvement in IDH1-deficient erythroblasts, ruling out oxidative stress as a driver .
IDH1 is highly expressed in male reproductive organs (e.g., testis, epididymis) and the liver . Its diagnostic utility lies in:
Glioma classification: Mutational status distinguishes IDH1-mutant gliomas from wild-type counterparts .
Therapeutic monitoring: D-2-HG levels in cerebrospinal fluid or blood correlate with treatment response .
IDH1 inhibitors target mutant enzymes to restore α-KG production and reduce D-2-HG.
IDH1 is a cytosolic NADP-dependent enzyme that catalyzes the oxidative decarboxylation of isocitrate to α-ketoglutarate (αKG). It is located in the cytoplasm and peroxisomes where it participates in lipid metabolism and glucose sensing . The normal enzyme functions as part of cellular metabolic pathways, with the wild-type protein maintaining critical roles in cellular redox status and energy production.
IDH1 functions through conformational changes between different states. Research has revealed an initial isocitrate (ICT)-binding state that transitions to a closed pre-transition state required for catalytic activity. This conformational change is essential for normal enzymatic function, with specific residues like Arg132 playing multiple functional roles in the catalytic reaction . Crystal structures of both wild-type and mutant IDH1 have provided significant insights into these mechanisms.
Human cells express three distinct IDH enzymes: cytosolic NADP-dependent IDH1, mitochondrial NADP-dependent IDH2, and mitochondrial NAD-dependent IDH3 . Unlike its mitochondrial counterparts, IDH1 is primarily cytosolic and uses NADP+ as a cofactor. This localization difference contributes to its distinct role in cellular metabolism.
The most prevalent tumor-specific mutation of IDH1 consists of a missense mutation at amino acid 132 that replaces an active-site arginine residue with histidine (R132H) . While R132H is the most common variant, other substitutions at this position can occur. These mutations are somatic and heterozygous, first discovered through genome-wide analysis of central nervous system tumors.
IDH1 mutations have been described in WHO Grade IV secondary glioblastomas (GBMs), WHO Grade II diffuse astrocytomas, WHO Grade II oligodendrogliomas, WHO Grade III anaplastic astrocytomas, WHO Grade III anaplastic oligodendrogliomas, and WHO Grade III anaplastic oligoastrocytomas . Approximately 70% of lower-grade gliomas harbor IDH1 mutations, making it a critical biomarker in neuro-oncology .
Mutant IDH1 has a neomorphic activity that converts α-ketoglutarate into 2-hydroxyglutarate (2HG) instead of the normal conversion of isocitrate to α-ketoglutarate . This altered enzyme consists of a dimer between wild-type and mutant proteins. The accumulated oncometabolite D-2-HG leads to epigenetic dysregulation, oncogenesis, and subsequent clonal expansion . Specifically, 2HG may inhibit various dioxygenases including PHD (prolyl hydroxylase), TET2, and histone demethylases, triggering aberrant angiogenesis and gene expression .
The R132H mutation hinders the conformational changes from the initial ICT-binding state to the pre-transition state, leading to impairment of the normal IDH activity . Additionally, the mutation renders a new reduction function that converts α-ketoglutarate to 2-hydroxyglutarate. Tyr139 has been shown to play a vital role in this gained reduction activity by compensating for the increased negative charge on the C2 atom of αKG during hydride anion transfer from NADPH .
Several methods have proven useful for diagnosing IDH1 mutation-bearing gliomas:
DNA genotyping of tumor specimens, involving sequencing of DNA extracted from brain tumor samples, which many clinical molecular diagnostic laboratories use to detect IDH1 and IDH2 mutations.
Immunohistochemical analysis using specific monoclonal antibodies targeting mutant IDH1, such as Imab-1, an anti-IDH1-R132H-specific antibody that reacts with mutated IDH1 .
Magnetic resonance spectrometry to detect 2HG, the downstream metabolite of mutant IDH1, which enables non-invasive molecular characterization of IDH1-mutated gliomas .
For optimal immunohistochemical detection, researchers should consider:
Selection of appropriate antibodies (e.g., Imab-1) that specifically recognize the R132H mutant protein.
Standardization of staining protocols, including fixation methods, antigen retrieval techniques, and detection systems.
Implementation of appropriate controls to validate staining specificity.
Analysis of staining patterns – notably, in immunohistochemical studies, every glioma cell in IDH1-positive samples stains positive for mutated IDH1, even along the infiltrating edge of the tumor into cortex .
Magnetic resonance spectrometry offers several unique advantages:
Non-invasive detection that doesn't require tissue sampling.
Ability to identify 2HG "hot spots" that can guide biopsy procedures.
Utility in assisting diagnostic workup and monitoring response to targeted therapies .
Limitations include technical challenges in distinguishing 2HG from other brain metabolites, variability in detection sensitivity, and the need for specialized equipment and expertise. Currently, no reports exist of increased 2HG in blood, urine, or cerebrospinal fluid of glioma patients harboring IDH1 mutations .
Several IDH1 inhibitors have been developed and evaluated clinically:
DS-1001: An oral brain-penetrant mutant IDH1 selective inhibitor that has shown efficacy in reducing D-2-HG levels and tumor size in preclinical studies .
Ivosidenib: An IDH1 inhibitor used for the treatment of IDH1-mutant gliomas, grades 2-4, which has demonstrated clinical benefit in both newly diagnosed and recurrent settings .
These inhibitors aim to reduce the production of the oncometabolite 2HG by targeting the neomorphic activity of mutant IDH1.
Clinical studies have shown promising results:
DS-1001: In a multicenter, open-label, dose-escalation phase I study for recurrent/progressive IDH1-mutant glioma, objective response rates were 17.1% for enhancing tumors and 33.3% for non-enhancing tumors. DS-1001 was well tolerated with favorable brain distribution .
Ivosidenib: Real-world data on ivosidenib treatment has shown concordance with controlled trials like INDIGO. The heterogeneous population evaluated in these studies provides insight into real-world IDH inhibitor use outside the strict confines of clinical trials .
Researchers evaluating IDH1 inhibitor response should consider:
Appropriate imaging protocols to assess tumor response, accounting for potential pseudoprogression.
Measurement of 2HG levels in tumors as a pharmacodynamic biomarker.
Standardized response criteria appropriate for gliomas.
Accounting for confounding factors such as concurrent treatments.
Distinguishing between prognostic effects (reflecting underlying disease characteristics) and predictive effects (truly reflective of response to therapy) .
Researchers can employ several sophisticated approaches:
X-ray crystallography to determine three-dimensional structures of wild-type and mutant IDH1 with and without substrates bound, as demonstrated in studies revealing the initial ICT-binding state and pre-transition state .
Site-directed mutagenesis coupled with biochemical assays to assess the functional impact of specific residues, such as the studies establishing Tyr139's role in the reduction activity of mutant IDH1 .
Molecular dynamics simulations to model protein dynamics and conformational changes that occur during catalysis.
Hydrogen-deuterium exchange mass spectrometry to map regions of conformational flexibility.
Researchers should consider multiple experimental systems:
Isogenic cell line models with wild-type and mutant IDH1 to isolate the specific effects of the mutation.
Patient-derived cell lines and xenografts that maintain the genetic background of IDH1-mutant tumors.
Genetically engineered mouse models expressing mutant IDH1 in relevant cell types.
Metabolomic profiling using mass spectrometry and NMR to comprehensively characterize metabolic alterations.
13C-labeled substrate tracing experiments to track metabolic flux through different pathways.
To investigate epigenetic changes induced by IDH1 mutations:
Chromatin immunoprecipitation sequencing (ChIP-seq) to map histone modification patterns.
Whole-genome bisulfite sequencing or reduced representation bisulfite sequencing to assess DNA methylation.
ATAC-seq to examine chromatin accessibility changes.
RNA-seq to correlate epigenetic alterations with gene expression changes.
Single-cell approaches to resolve cellular heterogeneity within tumors.
Functional assays to test the reversibility of epigenetic changes upon IDH1 inhibitor treatment.
Future IDH1 inhibitor development might focus on:
Structure-based drug design utilizing crystallographic data of the mutant enzyme to optimize binding and specificity.
Development of inhibitors that can target multiple IDH1 mutation variants beyond R132H.
Designing molecules with improved blood-brain barrier penetration for glioma treatment.
Creating dual inhibitors that can target both IDH1 and IDH2 mutations simultaneously.
Exploring combination strategies with other targeted therapies or standard treatments.
The neomorphic catalytic activities associated with IDH1 mutations may provide useful insights for metabolic engineering:
Knowledge of the structural changes that confer new catalytic activities can guide rational enzyme design.
Understanding the molecular basis for substrate specificity shifts can inform protein engineering strategies.
Mutant IDH1 has already contributed to the rapid design of enzymes for industrial applications, such as the large-scale production of adipic acid, a precursor in nylon synthesis .
This represents an example of how cancer-derived mutations can have broad applications beyond oncology research.
IDH1 is a soluble enzyme encoded by the IDH1 gene located on chromosome 2 in humans . The enzyme uses NADP+ as an electron acceptor, distinguishing it from other isocitrate dehydrogenases that use NAD+ . The human recombinant form of IDH1 is typically produced in E. coli and is available with a C-terminal 6-His tag for purification purposes .
Mutations in the IDH1 gene have been associated with various types of cancer, including gliomas and acute myeloid leukemia (AML). These mutations often result in a gain-of-function, leading to the production of an oncometabolite called 2-hydroxyglutarate (2-HG), which can contribute to tumorigenesis . IDH1 expression has also been correlated with poor survival in non-small cell lung cancer .
Recombinant human IDH1 is widely used in research to study its role in metabolism and cancer. It is also utilized in drug discovery efforts aimed at developing inhibitors for mutant IDH1 enzymes. The recombinant protein is typically supplied as a carrier-free formulation to avoid interference from other proteins, such as Bovine Serum Albumin (BSA) .
The recombinant human IDH1 protein is supplied as a 0.2 μm filtered solution in Tris, NaCl, Glycerol, Brij-35, and DTT. It is shipped with dry ice and should be stored at -70°C to maintain stability. The protein remains stable for six months from the date of receipt when stored at -70°C and for three months under sterile conditions after opening .