Dihydrofolate reductase (DHFR) is a critical enzyme in folate metabolism, catalyzing the reduction of dihydrofolate to tetrahydrofolate, a cofactor essential for purine and thymidine synthesis . The DHFR antibody is a research tool used to detect and study this enzyme in various biological contexts, including cancer, folate metabolism disorders, and therapeutic drug monitoring.
Detection of DHFR protein: Enables visualization of DHFR in tissues or cells via immunohistochemistry (IHC), immunofluorescence (IF), or Western blot (WB) .
Research applications: Investigates DHFR’s role in cancer progression, antifolate resistance, and gene expression regulation .
Therapeutic monitoring: Assesses DHFR levels in response to chemotherapeutics like methotrexate .
Purification: Protein-A affinity chromatography (monoclonal) or antigen affinity purification (polyclonal) .
Formulation: PBS buffer with glycerol and sodium azide for stability .
Antifolates (e.g., methotrexate, trimetrexate) increase DHFR protein levels in cancer cells by relieving translational repression . For example:
Human colon cancer cells: Exposure to methotrexate elevated DHFR-fused protein levels by 250-fold .
Xenograft models: Tumor DHFR levels increased 2–4-fold post-antifolate treatment, enabling positron-emission tomography (PET) imaging .
Molecular docking studies identified novel DHFR inhibitors with high affinity for the enzyme’s active site . Key findings:
Compound 16: Exhibited potent inhibition (IC50 < 10 nM) and structural similarity to methotrexate .
Resistance mechanisms: Chronic antifolate exposure induces DHFR gene amplification, a common resistance pathway .
Megaloblastic anemia: DHFR deficiency impairs folate metabolism, leading to anemia. Antibodies aid in diagnosing this condition .
Cancer biomarker: Elevated DHFR levels correlate with tumor aggressiveness and therapeutic resistance .
Methotrexate efficacy: DHFR antibody quantifies enzyme levels to predict drug response in cancer patients .
Gene therapy: DHFR-fused proteins are used to modulate gene expression in vivo, with antifolates enhancing their stability .
Methotrexate: A competitive DHFR inhibitor used in cancers like leukemia and breast cancer. Resistance often arises via DHFR gene amplification .
Targeted delivery: DHFR antibodies may guide antifolates to tumors, reducing systemic toxicity .
Dihydrofolate reductase (DHFR) is a key enzyme in folate metabolism that contributes to the de novo mitochondrial thymidylate biosynthesis pathway. It catalyzes an essential reaction for de novo glycine and purine synthesis, and for DNA precursor synthesis . DHFR is an established anti-cancer drug target whose inhibition disrupts folate metabolism and STAT3-dependent gene expression . As a ubiquitously expressed monomer considered to be a housekeeping gene, DHFR participates in multiple pathways across different cell types, making it a valuable target for both basic research and therapeutic development .
Human DHFR is a 21-23 kDa protein consisting of 187 amino acid residues with one DHFR domain (amino acids 4-185) . It exists in two pools within cells: one containing DHFR bound to its own RNA where it acts as a transcriptional repressor, and another containing DHFR bound to NADPH . The protein is localized in both mitochondria and cytoplasm . Its mRNA binding motif is suggested to involve Cys6, Leu22, Glu30, and Ser118, which can affect regulatory mechanisms .
Research-grade DHFR antibodies are available in several formats:
Different antibodies target specific regions of DHFR, including the N-terminus (amino acids 1-50) or regions surrounding particular residues such as Gly175 .
For optimal Western blot results with DHFR antibodies:
Sample preparation: Prepare whole cell lysates from established cell lines known to express DHFR (HeLa, Jurkat, COLO 205, HEK293T)
Loading amount: Load 20-30 μg of protein lysate per lane for adequate detection of endogenous DHFR
Blocking conditions: Use 5% non-fat dry milk in TBST as an optimal blocking buffer
Antibody dilutions: Dilutions range from 1:1000 to 1:10,000 depending on the specific antibody; monoclonal antibodies often perform well at higher dilutions (1:10,000)
Expected band size: Look for a specific band at approximately 21-22 kDa
Controls: Include positive controls such as recombinant DHFR protein at 0.1-0.2 μg
The experimental data shows that DHFR antibodies can detect the protein across multiple species including human, mouse, and rat samples due to high sequence homology (90% amino acid sequence identity) .
For successful IHC and ICC experiments:
Immunohistochemistry (IHC-P):
Antigen retrieval: Heat-mediated antigen retrieval in EDTA buffer (pH 8.0)
Blocking: 10% goat serum is recommended to reduce background
Antibody concentration: 2 μg/ml for paraffin-embedded sections
Incubation conditions: Overnight at 4°C for primary antibody
Detection systems: HRP-conjugated secondary antibodies with DAB chromogen work well
Validated tissues: Colorectal cancer tissue, small intestine, and colon show strong DHFR expression
Immunocytochemistry/Immunofluorescence (ICC/IF):
Cell fixation: Paraformaldehyde (4%) for cell fixation and permeabilization
Visualization: Use appropriate fluorophore-conjugated secondary antibodies (e.g., DyLight®550, NorthernLights™ 557)
Validated cell lines: U20S, MCF-7, PC-3 cells show reliable DHFR staining patterns
Modern techniques for measuring DHFR target engagement include:
Thermal Proteome Profiling (TPP): This technique helps confirm DHFR inhibitor binding in cellular contexts and can identify additional targets for inhibitors like methotrexate
DHFR accumulation assay: Measuring DHFR protein accumulation following inhibitor treatment serves as an indicator of target engagement
Biochemical assays coupled with cell-based assays: This combination provides a more comprehensive assessment of compound efficacy:
Research has shown that compounds like Cycloguanil and its analogues can engage DHFR in cells at sub-nanomolar concentrations, although growth impairments may not be observed until higher concentrations are reached .
Comprehensive validation should include:
Multiple detection methods: Cross-validate results using different techniques (WB, IHC, IF)
Species cross-reactivity testing: Validate across human, mouse, and rat samples due to the 90% sequence homology
Control experiments:
Knockout/knockdown validation: Compare antibody signal in DHFR-knockout or knockdown samples (like DG44 cells that lack DHFR)
Blocking peptide competition: Use the immunizing peptide to confirm signal specificity
Western blot data should show a clear band at the expected molecular weight of 21-22 kDa without significant non-specific banding .
Common challenges and solutions include:
Non-specific binding:
Epitope masking:
Variable expression levels:
Species cross-reactivity issues:
Signal detection sensitivity:
DHFR antibodies are valuable tools for investigating drug resistance mechanisms:
Protein expression analysis: Use Western blotting to quantify DHFR overexpression, which is directly associated with methotrexate (MTX) resistance. Studies have shown that cells with increased DHFR protein levels (5-fold higher) demonstrate significantly higher MTX resistance (IC₅₀ values of 1.03 μM vs 0.29 μM in wild-type cells)
SNP effects on DHFR expression: Investigate how SNPs in the DHFR gene affect antibody binding and protein levels. The 829C→T SNP near the miR-24 binding site in the 3′ UTR stabilizes DHFR mRNA, leading to 19-fold higher mRNA levels and 5-fold higher protein levels
miRNA regulation studies: Examine how miRNAs like miR-24 regulate DHFR expression by binding to the 3′ UTR. Combine with DHFR antibodies to assess protein levels after miRNA overexpression or inhibition
Target engagement analysis: Use DHFR antibodies in thermal shift assays to evaluate how mutations affect drug binding. This approach helps determine if resistance is due to reduced target binding or post-binding mechanisms
Subcellular localization changes: Investigate whether drug resistance correlates with altered subcellular distribution of DHFR between cytoplasmic and mitochondrial compartments
Cutting-edge applications include:
Companion diagnostics development: DHFR antibodies can be used to stratify patients based on DHFR expression levels, potentially predicting responsiveness to antifolate therapies
Polypharmacological studies: Recent research has illuminated that some DHFR inhibitors have additional targets. DHFR antibodies can help distinguish between on-target and off-target effects through target engagement assays
DHFR inhibitor screening:
Compensatory pathway investigation: Study how cancer cells adapt to DHFR inhibition by examining expression changes in related enzymes. For example, examining the relationship between DHFR inhibition and thymidylate synthase activity
Novel therapeutic strategy development: Recent work has shown that the antimalarial drug Pyrimethamine selectively inhibits human DHFR in cancer cell lines, which is linked to downstream inhibition of STAT3 signaling. DHFR antibodies are crucial for confirming the mechanism of action
Integrative approaches include:
CRISPR/Cas9 gene editing combined with antibody-based detection:
Proteomics and interactome studies:
Patient-derived xenograft (PDX) models:
Single-cell analysis techniques:
Computational biology integration:
Recent studies have revealed important interactions between DHFR and microRNAs:
miR-24 regulatory mechanism: DHFR antibodies have been instrumental in confirming that miR-24 regulates DHFR expression. When miR-24 binds to the 3′ UTR of DHFR mRNA, it leads to downregulation of DHFR protein. This regulation can be disrupted by SNPs like 829C→T, which interferes with miR-24 binding
mRNA half-life assessment: Studies using DHFR antibodies have shown that the 829C→T SNP increases DHFR mRNA half-life by 2-fold, contributing to protein overexpression and drug resistance
Therapeutic potential of miRNA mimics: Research suggests that miR-24 mimics may be valuable alone or in combination with methotrexate in treatment, with DHFR antibodies being crucial for monitoring the effectiveness of such approaches
SNP screening and personalized medicine: DHFR antibodies can help assess how various naturally occurring miRSNPs (SNPs located at or near microRNA binding sites) affect DHFR expression across different ethnic groups, potentially informing treatment outcomes and toxicity profiles
Combination therapy development: Understanding miRNA-mediated regulation of DHFR using antibody-based detection methods can lead to novel combination therapies targeting both the enzyme and its regulatory mechanisms
For optimal flow cytometry results with DHFR antibodies:
Cell preparation:
Blocking conditions:
Antibody concentration and incubation:
Controls:
Secondary antibody selection:
Data analysis considerations: