MTHFD2 is upregulated in multiple cancers and correlates with poor prognosis (Table 1).
Purine synthesis: Supplies 10-CHO-THF for de novo purine production, critical for DNA replication .
Redox regulation: Maintains NADPH pools, mitigating oxidative stress .
DNA repair: Binds PARP3 to enhance non-homologous end joining (NHEJ) in p53-deficient cancers .
MTHFD2 regulates T-cell function and inflammation (Table 2).
Th17/Treg balance: MTHFD2 depletion promotes Treg differentiation and suppresses Th17 pathogenicity .
mTORC1 signaling: AICAR accumulation (from MTHFD2 inhibition) downregulates mTORC1, impairing effector T-cell function .
MTHFD2 exhibits tissue-specific expression, particularly in immune and proliferative cells (Table 3).
MTHFD2 inhibition shows promise in oncology and immunology (Table 4).
Selectivity: MTHFD2 inhibitors must spare normal cells reliant on cytoplasmic MTHFD1 .
Resistance: Potential upregulation of MTHFD2L in response to inhibition .
MTHFD2 is a bifunctional mitochondrial enzyme with 5,10-methenyl-THF (CH+-THF) cyclohydrolase and 5,10-methylene-THF (CH2-THF) dehydrogenase activities . This 37kDa protein (350 amino acids) is primarily localized to the mitochondria and plays a crucial role in mitochondrial folate one-carbon metabolism .
Its enzymatic functions include:
Converting 5,10-methylene-THF to 5,10-methenyl-THF in the presence of NAD+ or NADP+
Catalyzing the cyclohydrolase reaction that converts 5,10-methenyl-THF to 10-formyl-THF
Methodologically, enzyme activity assays using purified recombinant MTHFD2 with varying concentrations of substrates and cofactors remain the gold standard for characterizing its catalytic properties. Researchers should consider using both forward and reverse reaction assays to fully understand its kinetic parameters.
MTHFD2 exhibits distinct expression patterns across different tissue states:
Developing tissues: Highly expressed during embryogenesis, particularly in early developmental stages, with expression beginning to taper off during later stages .
Normal adult tissues: Generally low or undetectable levels in most differentiated adult tissues .
Cancer tissues: Significantly upregulated in various cancer types and transformed cells .
Inflammatory conditions: Consistently overexpressed in multiple inflammatory and autoimmune diseases including ulcerative colitis, Crohn's disease, Celiac's disease, rheumatoid arthritis, systemic lupus erythematosus, psoriasis, psoriatic arthritis, Sjogren's syndrome, and multiple sclerosis .
To study expression patterns, researchers should employ multiple complementary techniques including RT-qPCR, Western blotting, immunohistochemistry, and RNA-seq. Careful selection of antibodies with confirmed specificity is essential, as is the inclusion of appropriate positive and negative controls.
MTHFD2 was initially characterized as strictly NAD+-dependent, but recent studies have revealed more complex cofactor utilization:
MTHFD2 can use both NAD+ and NADP+ as cofactors at physiologically relevant substrate concentrations .
This dual redox cofactor specificity is similar to that of MTHFD2L, which shares 60-65% amino acid sequence identity with MTHFD2 .
The relative use of NAD+ versus NADP+ has significant implications for the direction of one-carbon unit flux in mitochondria by affecting the equilibrium between 5,10-CH2-THF and 10-CHO-THF .
For experimental determination of cofactor specificity, researchers should conduct enzyme kinetics studies using purified enzyme with varying concentrations of both NAD+ and NADP+ under physiologically relevant conditions. Isothermal titration calorimetry and spectrophotometric assays can provide quantitative measurements of binding affinity and reaction rates with different cofactors.
MTHFD2 serves as a critical metabolic checkpoint controlling effector and regulatory T cell differentiation:
In activated T cells, MTHFD2 regulates de novo purine synthesis and signaling to promote proliferation and inflammatory cytokine production .
In pathogenic T helper-17 (Th17) cells, MTHFD2 prevents aberrant upregulation of the transcription factor FoxP3 and inappropriate gain of suppressive capacity .
MTHFD2 deficiency promotes regulatory T (Treg) cell differentiation .
MTHFD2 inhibition leads to depletion of purine pools, accumulation of purine biosynthetic intermediates, and decreased mTORC1 signaling .
MTHFD2 also regulates DNA and histone methylation in Th17 cells, suggesting epigenetic mechanisms of action .
Methodologically, researchers investigating MTHFD2's role in T cell biology should:
Use both genetic (CRISPR/Cas9) and pharmacological approaches to modulate MTHFD2 activity
Incorporate metabolic profiling (LC-MS) to measure purine metabolites and other relevant metabolic intermediates
Assess changes in mTORC1 signaling through phosphorylation status of downstream targets
Measure epigenetic modifications using techniques such as ChIP-seq and bisulfite sequencing
MTHFD2 exhibits several notable non-enzymatic "moonlighting" functions:
DNA damage repair: MTHFD2 unexpectedly promotes non-homologous end joining (NHEJ) in response to DNA damage by forming a complex with PARP3 to enhance its ribosylation .
PARP3 interaction: The introduction of a PARP3-binding but enzymatically inactive MTHFD2 mutant (D155A) is sufficient to prevent DNA damage, separating its enzymatic and non-enzymatic functions .
Nuclear localization: Despite being primarily a mitochondrial protein, MTHFD2 can localize to the nucleus where it may directly interact with DNA repair machinery .
To investigate these non-canonical functions, researchers should:
Use co-immunoprecipitation and proximity ligation assays to confirm protein-protein interactions
Employ subcellular fractionation and immunofluorescence microscopy to track MTHFD2 localization under different conditions
Design structure-function studies using point mutations that specifically disrupt enzymatic activity versus protein-protein interactions
Utilize DNA damage assays (comet assay, γH2AX foci formation) to assess functional outcomes
The p53-MTHFD2 axis represents a critical regulatory mechanism in cancer metabolism:
p53 transcriptionally suppresses MTHFD2 expression by binding to the MTHFD2 gene .
p53 loss or mutation leads to upregulation of MTHFD2, resulting in increased folate metabolism, de novo purine synthesis, and enhanced tumor growth both in vivo and in vitro .
MTHFD2 depletion strongly restrains proliferation in p53-deficient cells specifically and sensitizes these cells to chemotherapeutic agents .
MTHFD2 depletion in p53-deficient cells preferably induces apoptosis and cell proliferative arrest .
Conversely, in p53 wild-type cells, MTHFD2 downregulation can induce AICAR-mediated AMPK-p53-p21 activation, which may actually protect these cells .
Experimental approaches to study this relationship should include:
Chromatin immunoprecipitation (ChIP) to confirm p53 binding to the MTHFD2 promoter
Luciferase reporter assays to quantify transcriptional regulation
Isogenic cell line pairs (p53+/+ vs. p53-/-) to assess differential effects of MTHFD2 modulation
In vivo xenograft models with varying p53 status to validate therapeutic implications
MTHFD2 has significant prognostic implications across multiple cancer types:
Analyze large-scale cancer genomics databases (TCGA, ICGC) stratified by cancer type, stage, and molecular subtypes
Perform multivariate analyses to control for confounding factors
Validate findings using tissue microarrays and independent patient cohorts
Correlate MTHFD2 expression with other molecular markers to identify potential synergistic biomarkers
Researchers have several options for investigating MTHFD2 function through modulation:
Genetic approaches:
Pharmacological approaches:
Small molecule inhibitors targeting MTHFD2 enzymatic activity
Metabolic perturbations of related pathways (e.g., folate antagonists)
Combination approaches:
Rescue experiments with metabolic intermediates
Dual targeting of MTHFD2 and compensatory pathways
When designing MTHFD2 modulation experiments, researchers should consider:
The timing of intervention (e.g., before or after T cell activation)
The specificity of the approach (potential off-target effects)
The completeness of inhibition (partial vs. complete)
The cellular context (different cell types may respond differently)
Separating MTHFD2's dual roles requires specialized experimental designs:
Structure-function analysis:
Generate enzymatically inactive but structurally intact MTHFD2 mutants (e.g., D155A)
Compare phenotypes between wild-type, complete knockout, and catalytically inactive mutants
Metabolic rescue experiments:
Supplement with metabolic products (purines, one-carbon units) to bypass MTHFD2's enzymatic function
If phenotypes persist despite metabolic rescue, this suggests non-metabolic roles
Subcellular localization studies:
Generate constructs with altered localization signals (mitochondrial vs. nuclear)
Assess which functions correlate with specific localizations
Interactome analysis:
Identify protein interaction partners using techniques like BioID or IP-MS
Map interactions to specific functional domains of MTHFD2
These approaches should be complemented with comprehensive metabolomics and functional readouts to delineate which phenotypes result from metabolic versus non-metabolic activities.
MTHFD2 represents a promising target for inflammatory conditions:
MTHFD2 deficiency reduces disease severity in multiple in vivo inflammatory disease models .
MTHFD2 is consistently overexpressed across multiple inflammatory and autoimmune diseases including ulcerative colitis, Crohn's disease, and multiple sclerosis .
MTHFD2 expression is significantly elevated in recently diagnosed MS patients compared to healthy donors or MS patients undergoing therapy with disease remission .
MTHFD2 inhibition can promote regulatory T cell differentiation while impairing inflammatory T cell function .
Therapeutic development strategies should include:
High-throughput screening for selective MTHFD2 inhibitors
Testing in relevant animal models of inflammatory disease
Evaluation of effects on different immune cell subsets
Assessment of potential combination therapies with existing immunomodulatory drugs
Development of biomarkers to identify patients most likely to respond to MTHFD2-targeted therapy
Development of MTHFD2-targeted therapeutics faces several key challenges:
Selectivity: Achieving specificity over related enzymes like MTHFD2L and MTHFD1
Dual cofactor usage: Designing inhibitors effective against both NAD+ and NADP+-utilizing forms of the enzyme
Mitochondrial targeting: Ensuring sufficient drug penetration into mitochondria
Differential effects: Managing the opposing effects in p53-wild-type versus p53-deficient cancers
Resistance mechanisms: Identifying and addressing potential compensatory pathways
Researchers should approach these challenges through:
Structure-based drug design leveraging crystallographic data
Phenotypic screening in cancer models with defined p53 status
Development of mitochondria-targeted drug delivery systems
Combination strategies to address resistance mechanisms
Identification of biomarkers to select appropriate patients for MTHFD2-targeted therapy
The distinct temporal expression patterns of MTHFD2 and MTHFD2L during development present intriguing research opportunities:
MTHFD2 is expressed more abundantly during early developmental stages and begins to taper off, with little or no expression in most adult tissues .
MTHFD2L expression is low in early developmental stages but begins to increase at embryonic day 10.5 and remains elevated through birth and into adulthood .
This suggests a developmental switch from MTHFD2 to MTHFD2L to support one-carbon metabolism at different stages .
Future research directions should include:
Precise mapping of the spatiotemporal expression patterns of both enzymes during embryogenesis
Investigation of the regulatory mechanisms controlling this developmental switch
Functional studies using conditional knockout models with stage-specific deletion
Exploration of potential developmental defects resulting from altered expression timing
Assessment of the interplay between these enzymes and developmental signaling pathways
MTHFD2 appears to function at the intersection of multiple cellular stress response pathways:
DNA damage response: MTHFD2 directly participates in NHEJ repair through PARP3 interaction .
Metabolic stress: MTHFD2 influences AMPK-p53-p21 signaling when depleted in p53-competent cells .
Oxidative stress: MTHFD2 inhibition may lead to increased oxidative stress in cancer cells .
Immune activation: MTHFD2 expression is rapidly induced following T cell activation .
Future investigations should focus on:
Global interactome and phosphoproteome analysis of MTHFD2 under various stress conditions
Integrated multi-omics approaches to map MTHFD2-dependent responses to different stressors
Investigation of potential post-translational modifications regulating MTHFD2 function
Exploration of MTHFD2's role in cellular adaptation to microenvironmental stresses This comprehensive research agenda will help elucidate MTHFD2's position within cellular stress response networks and potentially identify novel therapeutic opportunities for conditions characterized by dysregulated stress responses.
Methylenetetrahydrofolate dehydrogenase (NADP+ dependent) 2, also known as MTHFD2, is a nuclear-encoded mitochondrial enzyme with bifunctional activities. It possesses both methylenetetrahydrofolate dehydrogenase and methenyltetrahydrofolate cyclohydrolase activities. This enzyme plays a crucial role in the folate metabolism pathway, which is essential for the synthesis of nucleotides and amino acids.
MTHFD2 functions as a homodimer and is unique in its absolute requirement for magnesium and inorganic phosphate . The enzyme is involved in the conversion of methylenetetrahydrofolate to methenyltetrahydrofolate, which is a critical step in the folate cycle. This cycle is vital for the production of purines and thymidylate, which are necessary for DNA synthesis and repair.
MTHFD2 is highly expressed in rapidly proliferating cells, such as cancer cells, and is considered a potential oncogene due to its strong association with poor prognosis and high levels of immune infiltrates in various cancers . The expression of MTHFD2 is regulated by several factors, including nutrient availability and cellular stress.
Recombinant human MTHFD2 is produced using Escherichia coli expression systems and is typically purified to a high degree of purity (>95%) suitable for various applications such as SDS-PAGE and mass spectrometry . The recombinant protein is often tagged with a His-tag at the N-terminus to facilitate purification and detection.
Recombinant MTHFD2 is used in research to study its role in folate metabolism, cancer biology, and potential as a therapeutic target. It is also utilized in biochemical assays to investigate its enzymatic activities and interactions with other proteins and small molecules.
Due to its elevated expression in cancer cells and its association with poor prognosis, MTHFD2 is being explored as a biomarker for cancer diagnosis and prognosis. Additionally, targeting MTHFD2 with specific inhibitors is being investigated as a potential therapeutic strategy for cancer treatment .