DHFR (dihydrofolate reductase) antibodies are immunoglobulins designed to specifically bind and detect the DHFR enzyme, a critical component in folate metabolism. DHFR catalyzes the reduction of dihydrofolate (DHF) to tetrahydrofolate (THF), a cofactor essential for purine synthesis, thymidylate generation, and DNA replication . These antibodies are engineered to recognize epitopes on the 21–23 kDa enzyme, which exists in two cellular pools: bound to its own RNA (acting as a transcriptional repressor) or to NADPH .
Clone PAT5B2AT (Prospec Bio):
MAB7934 (R&D Systems):
Rabbit IgG (Cell Signaling Technology #45710):
Rabbit IgG (Abcam ab49881):
DHFR antibodies enable precise detection and functional analysis of DHFR in diverse contexts.
Validation: Detects endogenous DHFR in human, mouse, and rat samples .
Example: Quantification of DHFR downregulation in cancer cells treated with 2-hydroxyoleic acid (HOA) .
HOA Treatment: Reduces DHFR mRNA/protein, inducing G₀/G₁ arrest in A549 cells and apoptosis in Jurkat cells .
Fusion Proteins: Antifolates (e.g., trimetrexate) upregulate DHFR-HSV1 TK fusion proteins, enhancing ganciclovir sensitivity in tumors .
| Source | Host | Reactivity | Sensitivity | Applications |
|---|---|---|---|---|
| Prospec Bio | Mouse | Human | Endogenous detection | WB, IP, ELISA |
| Cell Signaling | Rabbit | H, M, R, Mk | WB-validated | WB |
| Abcam | Rabbit | H, hamster | IP, WB, IHC-P | Recombinant fragment |
When using DHFR antibodies, optimal concentrations vary by application and specific antibody clone. For immunohistochemistry on formalin-fixed paraffin-embedded tissues, antibodies like ab124814 have been validated at 1/1000 dilution (0.20 μg/mL) with heat-mediated antigen retrieval using Tris-EDTA buffer (pH 9.0) . For Western blotting, typical working concentrations range from 0.5-2 μg/mL, with successful detection demonstrated using 1 μg/mL of anti-DHFR monoclonal antibody on PVDF membranes . For immunocytochemistry, higher concentrations are often required, with 10 μg/mL showing good results in MCF-7 cells with a 3-hour room temperature incubation .
Always perform an antibody titration experiment (using serial dilutions from 0.1-20 μg/mL) against positive control samples to determine the optimal concentration that maximizes signal-to-noise ratio for your specific experimental system.
DHFR antibodies vary in cross-reactivity across species. Several commercial antibodies detect human, mouse, and rat DHFR due to high sequence conservation. The MAB7934 antibody has validated reactivity with human, mouse, and rat DHFR in Western blot applications , while sc-377091 antibody is recommended for detection of DHFR from mouse, rat, and human origin .
To verify cross-reactivity:
Perform Western blot analysis using positive control lysates from each species of interest
Compare band sizes to the predicted molecular weight of DHFR (21 kDa)
Include negative controls (DHFR-knockdown samples if available)
Validate findings across multiple applications as cross-reactivity may differ between techniques
Use recombinant DHFR proteins from different species as standards
Cross-reactivity should be experimentally confirmed for each new application, as detection sensitivity may vary between species despite sequence homology.
Robust controls are critical for reliable DHFR antibody experiments:
Positive controls:
Cell lines with known DHFR expression (293T, Raji, C2C12, and Rat-2 have been validated)
Recombinant DHFR protein standards
Tissues with established DHFR expression patterns
Negative controls:
Primary antibody omission control
Isotype-matched non-specific antibody control
DHFR-knockdown or knockout samples when available
Peptide competition assays to confirm specificity
Technical controls:
Serial dilution of samples to confirm linear range of detection
For immunohistochemistry, include normal tissues with varying DHFR expression levels
For immunofluorescence, include autofluorescence controls
Implementing these controls allows proper interpretation of results and troubleshooting of unexpected findings across experimental systems.
DHFR exhibits dual localization in both the cytoplasm and mitochondria, with distribution patterns that may vary by cell type and physiological state . Immunofluorescence studies in MCF-7 human breast cancer cell lines show specific subcellular staining patterns that can be visualized using fluorescent-conjugated secondary antibodies and counterstaining with DAPI .
For accurate subcellular localization studies:
Use confocal microscopy for better resolution of compartments
Co-stain with mitochondrial markers (e.g., MitoTracker) and nuclear counterstains (DAPI)
Compare patterns across multiple cell types and tissue samples
Consider subcellular fractionation followed by Western blotting as a complementary approach
Use multiple DHFR antibody clones to confirm localization patterns
The subcellular distribution of DHFR may shift under certain conditions, including cell cycle progression, metabolic stress, or drug treatments targeting folate metabolism pathways.
Successful DHFR immunoprecipitation requires careful optimization:
Recommended lysis buffer composition:
Optimized procedure:
Prepare cell lysates on ice to preserve protein integrity
Pre-clear lysates with protein G-Sepharose to reduce non-specific binding
Incubate with anti-DHFR antibody (3-5 μg per mg total protein) at 4°C overnight
Add protein G-Sepharose 4 Fast Flow beads and incubate for 2-4 hours at 4°C
Wash beads 3-5 times with lysis buffer
Critical parameters:
Maintain samples at 4°C throughout to prevent protein degradation
Include negative controls (non-specific IgG, isotype control)
For studying DHFR complexes, consider milder detergents (0.5% NP-40)
Validate successful IP using Western blot with a different DHFR antibody clone
Antibodies specifically validated for immunoprecipitation, such as DHFR Antibody (A-9), will typically yield better results than those optimized solely for Western blotting .
When facing contradictory results from different DHFR antibody clones, implement this systematic troubleshooting approach:
Antibody characterization:
Determine epitopes recognized by each antibody
Assess specificity via Western blot in multiple cell lines (293T, Raji, C2C12, Rat-2)
Verify performance in knockout/knockdown validation studies
Review literature reports of each antibody's performance
Technical assessment:
Standardize sample preparation across experiments
Test multiple detection systems for each antibody
Evaluate buffer compatibility with each antibody
Consider effects of denaturing vs. native conditions on epitope accessibility
Biological considerations:
Different antibodies may recognize distinct DHFR isoforms
Post-translational modifications might affect epitope recognition
DHFR-protein complexes could mask certain epitopes
Resolution strategy:
Perform side-by-side comparison with standardized samples
Correlate antibody detection with DHFR activity assays
Consider using antibody combinations for comprehensive assessment
When publishing, clearly report which epitopes/antibodies were used
This structured approach transforms contradictory results into deeper insights about DHFR biology and regulation.
DHFR antibodies are valuable tools for investigating antifolate resistance mechanisms, particularly resistance to methotrexate (MTX):
Research applications:
Quantification of DHFR expression levels:
Western blot analysis comparing resistant vs. sensitive cells
Immunohistochemistry in tumor samples
Flow cytometry for single-cell analysis
Mutation-specific detection:
DHFR degradation kinetics:
DHFR-drug interaction studies:
Immunoprecipitation followed by drug binding assays
Antibody-based detection of conformational changes upon drug binding
Methodological framework:
Establish resistant cell lines through gradual MTX exposure
Compare DHFR expression using validated antibodies across multiple applications
Correlate protein expression with enzymatic activity measurements
Analyze cell cycle effects using propidium iodide staining and flow cytometry
Validate findings in patient-derived samples
This comprehensive approach enables mechanistic understanding of DHFR-mediated drug resistance and facilitates development of strategies to overcome treatment limitations.
Quantitative analysis of DHFR expression in immunohistochemistry requires standardized approaches:
Sample preparation:
Use consistent fixation protocols (10% neutral buffered formalin for 24-48 hours)
Perform heat-mediated antigen retrieval with Tris-EDTA buffer at pH 9.0
For antibodies like ab124814, use a dilution of 1/1000 (0.20 μg/mL)
Counterstain with hematoxylin for nuclear visualization
Scoring systems:
H-score (combines intensity and percentage of positive cells)
Allred score (useful for heterogeneous staining)
Automated image analysis using pixel-based quantification
Analytical parameters:
Staining intensity (negative, weak, moderate, strong)
Percentage of positive cells
Subcellular localization pattern
Spatial distribution within tissue architecture
Standardization approach:
Capture standardized digital images across all samples
Apply consistent thresholding parameters
Analyze multiple fields per sample (minimum 5-10 fields)
Include internal control tissues in each batch
Consider multi-observer scoring for validation
This systematic approach ensures reliable quantification and comparison of DHFR expression across different sample types and experimental conditions.
Multiplex immunofluorescence with DHFR antibodies allows simultaneous visualization of DHFR and other proteins:
Available conjugated antibodies:
FITC-conjugated (sc-377091 FITC)
PE-conjugated (sc-377091 PE)
Multiplexing strategies:
Sequential staining protocol:
Apply first primary antibody followed by fluorophore-conjugated secondary
Block remaining free binding sites
Apply DHFR antibody with differently-conjugated secondary antibody
Include nuclear counterstain (DAPI)
Direct conjugate approach:
Use directly conjugated DHFR antibodies
Combine with other directly conjugated antibodies with non-overlapping spectra
Optimize concentrations to achieve balanced signal intensities
Spectral unmixing:
Use specialized imaging systems capable of distinguishing overlapping fluorophores
Enable higher-order multiplexing (4+ targets simultaneously)
Optimization parameters:
Antibody concentration and incubation time
Order of antibody application
Blocking between sequential staining steps
Selection of compatible fluorophores with minimal spectral overlap
Controls for multiplex studies:
Single-stained controls for spectral compensation
Fluorescence-minus-one controls
Absorption controls to verify no cross-reactivity between antibodies
This approach allows visualization of DHFR in relation to other pathway components or cellular structures in a spatial context.
DHFR interacts with various proteins and even binds its own mRNA . Several antibody-based approaches can investigate these interactions:
Co-immunoprecipitation (Co-IP):
Use anti-DHFR antibodies to pull down DHFR and associated proteins
Perform Western blot analysis of co-precipitated partners
Consider mass spectrometry for unbiased identification of interactors
Optimal lysis conditions: 150 mM NaCl, 5 mM EDTA, 50 mM HEPES (pH 7.5), 1.0% Triton X-100
Proximity Ligation Assay (PLA):
Detect protein interactions in situ with <40 nm proximity
Use anti-DHFR antibody with antibodies against suspected partners
Visualize interactions as fluorescent spots via microscopy
Quantify interaction frequency using automated image analysis
FRET/BRET approaches:
Use fluorophore-conjugated DHFR antibodies (available as FITC, PE, Alexa Fluor conjugates)
Pair with differently labeled antibodies against interaction partners
Measure energy transfer as evidence of physical proximity
Optimize donor:acceptor ratios for maximum sensitivity
ChIP and RIP approaches:
Investigate DHFR interactions with nucleic acids
Particularly useful for studying DHFR's role in binding its own mRNA
Use highly specific DHFR antibodies validated for immunoprecipitation
These complementary approaches provide a comprehensive understanding of DHFR's protein interaction network in different cellular contexts.
Detection system selection significantly affects DHFR Western blot sensitivity and dynamic range:
Comparison of detection methods:
| Detection Method | Sensitivity | Dynamic Range | Multiplexing | Quantification |
|---|---|---|---|---|
| Chromogenic (HRP/DAB) | Low-Medium | ≈10-fold | No | Limited |
| Chemiluminescence | High | ≈100-fold | No | Good |
| Fluorescence | Medium-High | ≈1000-fold | Yes | Excellent |
| Near-infrared | High | ≈10000-fold | Yes | Excellent |
DHFR antibody conjugates available:
HRP-conjugated (sc-377091 HRP) for direct chemiluminescent detection
Various fluorophore conjugates for direct fluorescent detection
Optimization strategies:
For chemiluminescence:
Use high-sensitivity substrates for low abundance detection
Optimize exposure times (multiple exposures recommended)
Consider digital imaging for better dynamic range
For fluorescence:
Select appropriate wavelengths to minimize autofluorescence
Optimize scanning parameters (PMT gain, laser power)
Use compatible membrane types (low-fluorescence PVDF)
General considerations:
Block thoroughly to minimize background
Include positive controls at different dilutions
Use purified recombinant DHFR as a standard curve
For DHFR detection, successful Western blot has been demonstrated using PVDF membrane probed with 1 μg/mL of Mouse Anti-Human DHFR Monoclonal Antibody followed by HRP-conjugated secondary antibody, visualizing DHFR at approximately 21 kDa .
Variability in DHFR detection can stem from multiple sources that require systematic resolution:
Technical factors and solutions:
Antibody lot-to-lot variation:
Create master antibody stocks for long-term studies
Validate each new lot against previous standards
Consider site-specific recombinant antibody technology
Sample processing inconsistency:
Standardize lysis buffers and protein extraction protocols
Implement precise timing for all processing steps
Use automated systems where possible
Detection system variability:
Calibrate imaging equipment regularly
Include standard curves on each blot
Use digital imaging for better reproducibility
Biological factors and controls:
Cell cycle-dependent DHFR expression:
Growth conditions:
Standardize media composition and serum lots
Control temperature, CO₂, and humidity precisely
Document any deviations from standard conditions
Standardization approach:
Implement detailed protocol documentation
Use internal reference standards across experiments
Include quantitative loading controls appropriate for each experiment
Consider normalization to total protein methods
Implement statistical approaches appropriate for the experimental design
By systematically addressing these variables, researchers can achieve more consistent and reliable DHFR detection across independent experiments.
The "DHFR handle" system offers unique applications for fusion protein studies. Based on research by Iwakura et al., this system can be applied immunologically for small peptides like leucine enkephalin (LEK) :
Principles of the DHFR handle system:
DHFR serves as a protein fusion partner ("handle")
Small peptides fused to DHFR maintain their conformational integrity
The system enables large-scale preparation of otherwise difficult-to-produce peptides
DHFR antibodies can be used to detect, purify, or track the fusion proteins
Experimental approach:
Generate DHFR-peptide fusion constructs
Express and purify using standard recombinant protein techniques
Use anti-DHFR antibodies for:
Western blot detection of fusion proteins
Immunoprecipitation of fusion complexes
Immunoaffinity purification
Immunofluorescence localization studies
Validation methods:
Competitive binding assays to assess epitope accessibility
Verification that DHFR antibodies recognize the handle portion without interfering with peptide function
IC₅₀ value determination for competitive binding (e.g., LEK showed IC₅₀ values of 3.74×10⁻⁶ and 4.66×10⁻⁶ M with two different monoclonal antibodies)
This approach is particularly valuable for studying small bioactive peptides and protein fragments that are otherwise challenging to work with due to size, stability, or detection limitations.
DHFR expression varies significantly across cancer cell lines, requiring careful experimental design:
Cell line selection considerations:
Include both methotrexate-sensitive and resistant lines
Select lines representing diverse tissue origins
Consider lines with known DHFR gene amplification status
Include normal cell counterparts for comparison
Validated cell lines for DHFR studies:
293T human embryonic kidney cells
Raji human Burkitt's lymphoma cells
MCF-7 human breast cancer cells
C2C12 mouse myoblast cells
Experimental standardization:
Maintain consistent culture conditions across all lines
Harvest cells at similar confluence (70-80%)
Use multiple DHFR antibodies targeting different epitopes
Correlate protein expression with mRNA levels and enzyme activity
Advanced analysis approaches:
Compare subcellular localization patterns across cell types
Assess response to folate pathway perturbations
Correlate DHFR expression with proliferation rates and cell cycle profiles
Consider drug sensitivity testing in relation to DHFR levels
By systematically characterizing DHFR expression across diverse cancer cell lines, researchers can gain insights into the relationship between DHFR expression, cell type specificity, and potential therapeutic targeting strategies.
Differentiating between DHFR isoforms requires specialized antibody-based strategies:
Background on DHFR isoforms:
Alternative splicing yields multiple DHFR isoforms
The canonical human DHFR protein has 187 amino acid residues and a mass of 21.5 kDa
Antibody selection strategies:
Isoform-specific antibodies:
Select antibodies targeting unique regions in specific isoforms
Validate specificity using recombinant isoform proteins
Consider custom antibody development for poorly characterized isoforms
Differentiation by molecular weight:
Use high-resolution SDS-PAGE to separate closely related isoforms
Western blot with antibodies recognizing common epitopes
Compare to predicted molecular weights of known isoforms
2D gel electrophoresis approach:
Separate isoforms by both isoelectric point and molecular weight
Immunoblot to identify DHFR-specific spots
Perform mass spectrometry on isolated spots for definitive identification
Immunoprecipitation with isoform discrimination:
Use isoform-specific antibodies for selective precipitation
Analyze precipitated proteins by mass spectrometry
Confirm differential interactions with binding partners
This methodological approach enables researchers to distinguish between closely related DHFR isoforms and study their specific functions in different cellular contexts.
Correlating DHFR antibody detection with enzymatic activity provides crucial validation:
Enzymatic activity measurement:
The specific activity of DHFR can be determined through established protocols
Typical assays monitor the conversion of dihydrofolate to tetrahydrofolate with NADPH as cofactor
Spectrophotometric methods track NADPH oxidation at 340 nm
Results can be expressed as nmol substrate converted per minute per mg protein
Correlation methodologies:
Parallel analysis:
Process identical samples for both antibody detection and activity assays
Plot quantitative antibody signal versus specific activity
Calculate correlation coefficients and regression analysis
Cell-based approaches:
Use cell lines with varying DHFR expression levels
Perform immunocytochemistry for spatial DHFR distribution
Correlate with enzymatic activity in cell lysates
Include MTX-resistant variants with altered DHFR properties
Manipulation-based validation:
In situ approaches:
Develop histochemical assays for DHFR activity
Compare spatial patterns with immunohistochemistry results
Use dual labeling where feasible
This integrated approach validates antibody performance while providing insights into the relationship between DHFR protein levels and functional activity across different experimental conditions.
Recent technological advances are expanding the utility of DHFR antibodies in research settings:
Super-resolution microscopy:
Enables visualization of DHFR at the nanoscale level
Reveals previously undetectable subcellular distribution patterns
Requires highly specific antibodies with minimal background
Mass cytometry (CyTOF):
Uses metal-conjugated antibodies for highly multiplexed cellular analysis
Allows simultaneous detection of DHFR with 40+ other proteins
Eliminates fluorescence spectrum limitations
Proximity-dependent biotinylation:
DHFR-BioID or DHFR-APEX2 fusion proteins
Maps protein interaction networks in living cells
Can be detected with both anti-DHFR and anti-biotin antibodies
Single-cell proteomics:
Measures DHFR levels in individual cells within heterogeneous populations
Correlates with other protein markers and cellular phenotypes
Requires highly sensitive and specific antibodies
Antibody engineering:
Recombinant antibody technology for consistent performance
Site-specific conjugation for improved sensitivity
Nanobodies and single-chain antibodies for improved penetration
These emerging technologies are expanding the capabilities of DHFR antibody applications across multiple research domains, from basic biochemistry to translational medicine.
When publishing DHFR antibody-based research, address these critical methodological considerations:
Comprehensive antibody reporting:
Validation documentation:
Specificity controls (knockout/knockdown, blocking peptide)
Cross-reactivity assessment for relevant species
Application-specific validation (not assuming WB validation transfers to IHC)
Lot-specific validation if using different lots
Inclusion of positive and negative controls
Detailed methodological protocols:
Complete sample preparation procedures
Buffer compositions and reaction conditions
Incubation times and temperatures
Detection systems with complete parameters
Image acquisition settings and analysis parameters
Quantification and statistics:
Clear description of quantification methods
Number of biological and technical replicates
Appropriate statistical tests with justification
Effect size reporting alongside p-values
Raw data availability statement
Dihydrofolate reductase (DHFR) is a crucial enzyme involved in the metabolic processes of cells. It plays a significant role in the synthesis of nucleotides, which are the building blocks of DNA. The enzyme is found in all organisms, from bacteria to humans, and is essential for cell growth and proliferation .
DHFR catalyzes the reduction of dihydrofolate to tetrahydrofolate using NADPH as an electron donor . Tetrahydrofolate is a key cofactor in the synthesis of purines, thymidylic acid, and certain amino acids. This reaction is vital for the de novo synthesis of nucleotides, which are necessary for DNA replication and repair .
In humans, the DHFR gene is located on chromosome 5 at the q14.1 region . The gene has multiple intronless processed pseudogenes or DHFR-like genes identified on separate chromosomes . Mutations or deficiencies in DHFR can lead to megaloblastic anemia, a condition characterized by the presence of large, abnormal red blood cells .
There are two structural classes of DHFR:
Mouse anti-human DHFR antibodies are monoclonal antibodies produced by immunizing mice with human DHFR protein. These antibodies are used in various research applications, including: