Human dihydroorotate dehydrogenase (DHODH) is a mitochondrial enzyme critical for the de novo pyrimidine biosynthesis pathway, catalyzing the oxidation of dihydroorotate to orotate using ubiquinone as an electron acceptor . As a class 2 DHODH, it is membrane-bound, distinguishing it from cytosolic class 1 variants . DHODH’s role in nucleotide synthesis makes it a therapeutic target for autoimmune diseases (e.g., rheumatoid arthritis) and cancers . Approved inhibitors like teriflunomide (a metabolite of leflunomide) highlight its clinical relevance, though limitations in safety and efficacy drive ongoing research .
DHODH converts dihydroorotate to orotate, a rate-limiting step in pyrimidine synthesis . Its inhibition disrupts nucleotide pools, inducing ribosomal stress and cell cycle arrest, particularly in rapidly proliferating cells like cancer . In autoimmune conditions, reduced pyrimidine availability suppresses lymphocyte activation .
Teriflunomide and brequinar act via distinct binding modes, with teriflunomide stabilizing a closed FMN conformation .
Compound 1289/1291 (6-isopropyl-benzo[d] triazol-4-one derivatives) exploit hydrophobic pockets, achieving sub-10 nM IC₅₀ values .
HZ05 enhances p53 activation by inhibiting DHODH, linking pyrimidine stress to tumor suppression .
Autoimmune Diseases: Teriflunomide reduces lymphocyte proliferation by depleting pyrimidines, approved for multiple sclerosis and rheumatoid arthritis .
Cancer: DHODH inhibition induces S-phase arrest and ribosomal stress, particularly effective in acute myeloid leukemia (AML) . Compound 3 (ChemBridge) shows promise with cellular IC₅₀ values ≤2.3 μM .
Synergistic Effects: Combining DHODH inhibitors with brequinar enhances immunosuppressive activity in autoimmune models .
Selectivity and Safety: Existing inhibitors (e.g., teriflunomide) face narrow therapeutic windows due to off-target effects .
Structure-Based Design: High-resolution crystallography (e.g., 1.6 Å structures of DHODH-1289 complexes) reveals binding motifs, enabling scaffold hopping .
Natural Product Screening: Phytoconstituents from immunomodulatory plants (e.g., Withania, Curcuma) show potential as DHODH inhibitors, offering cost-effective alternatives .
Targeted DHODH Inhibitors: Nanomolar inhibitors (e.g., HZ05, 1289) with improved selectivity are advancing into preclinical trials .
Combination Therapies: Pairing DHODH inhibitors with immunomodulators (e.g., brequinar + plant extracts) may enhance efficacy .
Natural Product Derivatives: Phytochemicals offer untapped potential for safer, cost-effective DHODH inhibitors .
p53 Activation: DHODH inhibitors like HZ05 leverage pyrimidine stress to activate p53, a novel strategy for tumor suppression .
Dihydroorotate dehydrogenase (DHODH) is a mitochondrial enzyme that plays a crucial role in the de novo pyrimidine biosynthesis pathway. It catalyzes the rate-limiting step in this pathway, specifically the conversion of dihydroorotate to orotate. This reaction is essential for the subsequent production of pyrimidines, which are fundamental building blocks of DNA and RNA .
Methodologically, researchers studying DHODH function typically employ enzymatic assays measuring the conversion of dihydroorotate to orotate, often using spectrophotometric methods that track changes in absorbance associated with the reduction of electron acceptors.
DHODH is primarily localized in the mitochondria of human cells. Specifically, it is found in the inner mitochondrial membrane where it participates in the fourth step of pyrimidine biosynthesis . This localization is significant because it enables the enzyme to connect pyrimidine synthesis to the mitochondrial electron transport chain.
To study DHODH localization, researchers commonly use subcellular fractionation techniques followed by Western blotting, or fluorescence microscopy with antibodies specific to DHODH. The enzyme's association with the mitochondrial membrane means that proper solubilization techniques are essential when isolating it for functional studies.
Several variants in the DHODH gene have been identified as causative for Miller syndrome, a rare condition affecting facial, arm, and leg development. These variants typically result in amino acid substitutions in the dihydroorotate dehydrogenase enzyme .
Miller syndrome appears to disrupt the development of the pharyngeal arches, which are paired structures that form during embryonic development and ultimately develop into the bones, nerves, and muscles of the head and neck. Although DHODH is active in these structures during development, the exact mechanism by which DHODH variants lead to the specific facial features of Miller syndrome remains unclear .
Additionally, the bones of the arms and legs often develop abnormally in people with Miller syndrome. While DHODH appears to be active in the limb buds during early development, the precise relationship between DHODH variants and the specific bone abnormalities observed is still being investigated .
DHODH expression has been identified as an independent unfavorable prognostic marker in neuroblastoma. High DHODH expression significantly correlates with poor survival across all stages of the disease. Particularly, the highest levels of DHODH expression are observed in high-risk MYCN-amplified tumors, and expression increases with disease stage .
Methodologically, researchers can assess DHODH expression using RNA sequencing or quantitative PCR for mRNA levels, or immunohistochemistry for protein expression. Kaplan-Meier survival analyses with stratification based on DHODH expression levels can help establish its prognostic value in neuroblastoma patient cohorts.
Several classes of DHODH inhibitors have been developed and are being researched:
Established clinical inhibitors:
Novel synthetic inhibitors:
Natural product inhibitors:
For researchers, selection of an appropriate inhibitor depends on the specific research question, desired potency, and pharmacokinetic/pharmacodynamic requirements.
High-resolution crystal structures have revealed detailed interactions between DHODH inhibitors and the enzyme. Human DHODH has an α/β-barrel fold, and inhibitors bind at a tunnel within the enzyme structure together with FMN .
Compounds like 1289 and 1291 are stabilized by numerous hydrophobic interactions involving specific amino acid residues including M43, L46, L58, F62, F98, M111, and L359 . The DHODH inhibitor binding site demonstrates intrinsic plasticity, allowing it to accommodate diverse inhibitor scaffolds, which explains why structurally different molecules can effectively inhibit the enzyme.
The crystal structures of human DHODH complexed with various inhibitors provide valuable insights for structure-based drug design. Key methodological approaches include X-ray crystallography (as shown in the table below), molecular docking studies, and structure-activity relationship analysis.
Parameter | DHODH-1289 Complex | DHODH-1291 Complex |
---|---|---|
Resolution (Å) | 1.75 | 1.90 |
R work/ R free | 0.157/0.176 | 0.168/0.195 |
Number of Protein Atoms | 2,768 | 2,821 |
Number of Ligand/Ion Atoms | 155 | 112 |
Number of Water Molecules | 330 | 282 |
Table 1: Crystallographic data for DHODH-inhibitor complexes
For evaluating DHODH inhibitors in autoimmune disease models, researchers employ several methodological approaches:
In vitro assessment:
Enzymatic kinetic analysis to determine inhibitory potency
Thermofluor assays to assess binding affinity through thermal stabilization
Isothermal titration calorimetry for quantitative binding measurements
Concanavalin A-triggered T-cell assays to assess immunomodulatory effects
Mixed lymphocyte reaction assays to evaluate impact on immune cell proliferation
In vivo assessment in experimental autoimmune models:
MOG-induced experimental allergic encephalomyelitis (EAE) in mice as a model for multiple sclerosis
In vivo imaging to evaluate myelin destruction using specific dyes (e.g., DBT dye administered intravenously)
Blood-brain barrier integrity assessment
Histochemical analysis of tissue sections using hematoxylin and eosin (H&E) staining and luxol fast blue (LFB) staining
The natural product piperine has been evaluated using these methodologies and identified as a DHODH inhibitor with potential therapeutic effects in experimental models of multiple sclerosis .
Multi-omic approaches have proven valuable for identifying DHODH as a metabolic vulnerability in cancers like neuroblastoma. These approaches integrate:
Metabolomics: Analysis of metabolite profiles revealed that neuroblastoma cell lines accumulate specific metabolites compared to other solid tumor cell lines, including pyrimidines (2-deoxycytidine and cytidine), the purine derivative hypoxanthine, and glutamine, which provides nitrogen for nucleotide synthesis .
Transcriptomics: Gene expression analysis across tumor types showed that DHODH is expressed at high levels in neuroblastoma and rhabdoid tumors compared to other solid tumors and pediatric samples .
Functional genomics: CRISPR screening data from the Cancer Cell Line Encyclopedia (CCLE) identified a correlation between DHODH dependency and MYCN dependency in neuroblastoma cell lines .
Partial least-squares discriminant analysis: This statistical approach can be used to identify metabolites that specifically accumulate in certain cancer types, such as neuroblastoma .
For researchers, integration of these multi-omic datasets requires sophisticated computational approaches and can provide comprehensive insights into the metabolic dependencies of cancer cells that would not be apparent from any single analysis method.
Research has revealed an important relationship between DHODH dependency and cellular differentiation states, particularly in neuroblastoma. Key methodological approaches include:
Transcriptional profiling to classify cell lines based on differentiation status:
Correlation analysis between differentiation status and DHODH dependency:
DHODH CERES scores (a measure of gene dependency from CRISPR screens) can be compared across cell lines with different differentiation states
Neuroblastoma cell lines with mesenchymal features (high MES scores) typically show lower DHODH dependency
Conversely, cell lines most dependent on DHODH generally have low MES scores
Sensitivity testing of differentiated cells to DHODH inhibitors:
These findings suggest that cellular differentiation state is an important factor to consider when evaluating DHODH as a therapeutic target in cancer.
Combination approaches involving DHODH inhibitors and standard chemotherapeutics have shown promising results in preclinical models. Key methodological considerations include:
In vitro combination studies:
Dose-response matrices testing various concentrations of DHODH inhibitors and chemotherapeutics
Cell viability/cytotoxicity assays to quantify therapeutic effects
Calculation of combination indices to determine synergy, additivity, or antagonism
In vivo combination studies:
Mechanistic investigations of combination effects:
Analysis of cell cycle effects
Assessment of DNA damage and repair mechanisms
Evaluation of changes in pyrimidine metabolism that might sensitize cells to chemotherapy
Biomarker identification for combination therapy response:
These methodological approaches are critical for translating promising preclinical findings with DHODH inhibitor combinations into clinical applications.
Translating DHODH inhibitor research into clinical applications faces several challenges that researchers must address:
Target specificity and off-target effects:
Developing highly selective DHODH inhibitors to minimize off-target effects
Differentiating between effects due to DHODH inhibition versus other mechanisms
Employing rescue experiments with pyrimidine supplementation to confirm on-target activity
Biomarker development:
Identifying and validating predictive biomarkers of response
In neuroblastoma, high DHODH expression identifies a "highest risk group" that might benefit most from DHODH inhibition
Determining whether DHODH expression alone or in combination with other markers (like MYCN status) best predicts response
Resistance mechanisms:
Optimal dosing and scheduling:
Determining optimal dose and schedule for DHODH inhibitors, particularly in combination regimens
Minimizing toxicity while maintaining efficacy
Considering pharmacokinetic and pharmacodynamic relationships
These challenges require rigorous preclinical investigation using appropriate disease models and careful clinical study design to advance DHODH inhibitors as therapeutic agents.
Artificial intelligence (AI) and computational methods offer promising approaches to accelerate DHODH inhibitor discovery:
Structure-based drug design:
Quantitative structure-activity relationship (QSAR) modeling:
Developing predictive models based on known DHODH inhibitors and their potencies
Identifying key molecular features that contribute to DHODH inhibitory activity
Utilizing these models to optimize lead compounds
Molecular dynamics simulations:
Systems biology approaches:
Modeling the impact of DHODH inhibition on pyrimidine metabolism in different cellular contexts
Predicting combination strategies that might synergize with DHODH inhibition
Identifying potential biomarkers of response to DHODH inhibition
These computational approaches can significantly reduce the time and resources required for traditional drug discovery methods while increasing the likelihood of identifying effective DHODH inhibitors.
Advancing our understanding of DHODH in development and disease will benefit from several novel experimental models:
Patient-derived organoids:
Three-dimensional cultures derived from patient tumors that better recapitulate tumor heterogeneity
Evaluation of DHODH inhibitor response across organoids derived from different patients
Correlation of organoid response with patient characteristics and outcomes
Developmental models for Miller syndrome:
Single-cell approaches:
Single-cell RNA sequencing to assess heterogeneity in DHODH expression within tumors
Spatial transcriptomics to understand DHODH expression in the context of tissue architecture
Single-cell metabolomics to evaluate cell-to-cell variation in pyrimidine metabolism
Humanized mouse models:
Mice with human immune systems for studying DHODH inhibition in autoimmune conditions
Evaluation of both therapeutic efficacy and potential immune-related adverse effects
These experimental models will provide more nuanced insights into DHODH biology and facilitate the translation of basic research findings into clinical applications.
Dihydroorotate dehydrogenase (DHODH) is a crucial enzyme in the de novo synthesis pathway of pyrimidine nucleotides, which are essential for the synthesis of DNA and RNA. This enzyme is particularly significant in rapidly proliferating cells, such as cancer cells, due to their high demand for nucleotides .
DHODH is a flavin-dependent enzyme located on the outer surface of the inner mitochondrial membrane . It catalyzes the oxidation of dihydroorotate to orotate, a key step in the pyrimidine biosynthesis pathway . The human DHODH enzyme is a monomeric protein that belongs to Class 2 of the DHODH family . It is ubiquitously expressed in most tissues .
Recombinant human DHODH is produced using E. coli expression systems. The recombinant protein typically includes an N-terminal His-tag to facilitate purification . The recombinant form retains the enzymatic activity of the native protein, making it useful for various research applications, including drug discovery and biochemical studies .
DHODH has emerged as a promising therapeutic target for several diseases. Initially, it was targeted for the treatment of non-neoplastic diseases such as rheumatoid arthritis and multiple sclerosis . More recently, DHODH inhibitors have shown potential in cancer therapy due to the enzyme’s role in nucleotide synthesis and mitochondrial function .
Recombinant human DHODH is widely used in research to study its role in cellular metabolism and disease. It is also employed in high-throughput screening assays to identify potential DHODH inhibitors . These inhibitors could serve as therapeutic agents for diseases characterized by rapid cell proliferation, such as cancer .