DHFR Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to DHFR Antibody

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.

Key Functions of DHFR Antibody

  • 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 .

2.2. Immunogen and Purification

  • Immunogen: Recombinant human DHFR protein (aa 1-187) .

  • Purification: Protein-A affinity chromatography (monoclonal) or antigen affinity purification (polyclonal) .

  • Formulation: PBS buffer with glycerol and sodium azide for stability .

3.1. Antifolate-Induced DHFR Upregulation

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 .

3.2. DHFR Inhibitors and Resistance

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 .

4.1. Diagnostic Use

  • 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 .

4.2. Therapeutic Monitoring

  • 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 .

5.1. Cancer Therapy

  • 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 .

5.2. Infectious Diseases

  • Antimicrobial agents: DHFR inhibitors like trimethoprim target bacterial enzymes, avoiding mammalian DHFR due to structural differences .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can dispatch products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery times.
Synonyms
DHFR antibody; DHFRP1 antibody; Dihydrofolate reductase antibody; DYR antibody; DYR_HUMAN antibody; EC 1.5.1.3 antibody
Target Names
DHFR
Uniprot No.

Target Background

Function
DHFR (Dihydrofolate reductase) plays a pivotal role in folate metabolism. It contributes to the de novo mitochondrial thymidylate biosynthesis pathway. DHFR catalyzes an essential reaction for de novo glycine and purine synthesis, as well as for DNA precursor synthesis. It binds to its own mRNA and that of DHFR2.
Gene References Into Functions
  1. In patients with Acute Lymphoblastic Leukemia (ALL) and Non-Hodgkin Lymphoma (NHL) undergoing methotrexate treatment, an evaluation of treatment toxicities and outcomes was conducted. Multivariate analysis revealed that the DHFR-1610G/T (OR=0.107, p=0.018) and MTHFR677T alleles (OR=0.12, p=0.026) exhibited a strong protective effect against hematologic toxicity. Conversely, the DHFR-1610CC genotype was associated with increased hematologic toxicity (OR=9, p=0.045). PMID: 28887233
  2. Dihydrofolate reductase and thymidylate synthase form a complex in vitro and co-localize in both normal and cancerous cells. PMID: 27187663
  3. Single nucleotide polymorphisms within the DHFR gene have been linked to Systemic Lupus Erythematosus. PMID: 28943344
  4. A study concluded that the 63/91 polymorphism in the DHFR gene promoter can modulate the onset of methotrexate-related adverse effects in rheumatoid arthritis patients. PMID: 27636122
  5. Findings suggest that identifying DHFR polymorphisms in the promoter region of the gene may be beneficial in tailoring methotrexate doses for pediatric ALL patients on maintenance therapy. PMID: 28719513
  6. The abundance of dihydrofolate reductase was significantly higher in biopsies from rheumatoid arthritis (RA) patients compared to controls. This increased expression correlated with the dosage of methotrexate (MTX), the most commonly prescribed immunosuppressive drug for RA. PMID: 27627584
  7. This study demonstrated that ADAR1 positively regulates DHFR expression by editing the miR-25-3p and miR-125a-3p binding sites in the 3'-UTR of DHFR, enhancing cellular proliferation and resistance to methotrexate in MCF-7 cells. PMID: 28188287
  8. The findings suggest that folate nutrition and the 19bp del-DHFR [Dihydrofolate reductase] variation may interact to modify the risk of adenomatous polyps, a precursor to colorectal cancer. PMID: 26875486
  9. The highest expression of GGH and EGFR was observed in the left-sided colon, while the highest expression of DHFR, FPGS, TOP1, and ERCC1 was noted in the rectosigmoid. TYMP expression was approximately equivalent in the right-sided colon and rectum. PMID: 26676887
  10. Overexpression of miR-192 inhibited cellular proliferation by binding to DHFR. miR-192 also decreased cellular anchoring via repression of ITGAV, ITGB1, ITGB3, and CD47. PMID: 26506238
  11. Data suggest that DHFR exhibits intrinsic activity kinetics that are temperature-independent. Additionally, increased mass (e.g., incorporation of H, C, and N isotopes) has no effect on intrinsic activity kinetics or the protein conformation/stability of DHFR. PMID: 26813442
  12. Patients homozygous for the G allele of rs1053129 in the DHFR gene were more likely to experience metastasis (45%, P= 0.005). Additionally, the methylenetetetrahydrofolate reductase (MTHFR) 677C allele was associated with a higher degree of liver toxicity. PMID: 25778468
  13. This study examined the association between cognitive outcomes, the 19-bp deletion DHFR polymorphism, folate status, and their interaction with high or normal plasma folate levels. PMID: 26354538
  14. S-nitrosylation of DHFR at cysteine 7 by eNOS-derived NO is crucial for DHFR stability. PMID: 26381869
  15. Genetic association studies conducted in a cohort of healthy young adults in Ireland revealed that a 19 bp deletion/insertion polymorphism within intron 1 of DHFR (rs70991108) is not associated with folate nutritional status in the studied population. PMID: 26269242
  16. Dihydrofolate Reductase and Thymidylate Synthase Transgenes Resistant to Methotrexate Interact to Permit Novel Transgene Regulation. PMID: 26242737
  17. Results from this study, for the first time, suggested that the DHFR 19-bp D/D genotype may confer a reduced risk of Non-Syndromic Cleft Lip with or without Cleft Palate (NS-CL/P) and could act as a protective factor against NS-CL/P in Iranian subjects. PMID: 26221921
  18. Triple mutant haplotypes AIRNI (N51I+C59R+S108N) of the dhfr gene, associated with pyrimethamine resistance, were prevalent in the Chirang district of Assam. PMID: 25511211
  19. Human dihydrofolate reductase is relatively less stable than its E.coli counterpart. PMID: 26349210
  20. Subpicosecond protein motion is dynamically coupled to hydride transfer catalyzed by hsDHFR but not ecDHFR. PMID: 25369552
  21. Genetic variations in MTHFR, DHFR, and ATIC can be considered pharmacogenetic markers of treatment outcome in RA patients under methotrexate monotherapy. PMID: 25084201
  22. Genome-wide association studies have identified 10 novel genetic loci as risk factors for methotrexate response in rheumatoid arthritis patients, in addition to polymorphisms in DHFR, FPGS, and TYMS. PMID: 24583629
  23. Genetic association between DHFR single nucleotide polymorphisms and nonsyndromic cleft lip with or without cleft palate. PMID: 24361572
  24. Human DHFR requires minimal backbone conformational rearrangement as it proceeds through its enzymatic cycle, but ligand flux is mediated by more subtle conformational changes that depend on the side chain motions of critical residues. PMID: 24498949
  25. Berberine suppresses the growth of cisplatin-resistant cells more effectively than sensitive counterparts by interfering with the expression of folate cycle enzymes, dihydrofolate reductase (DHFR) and thymidylate synthase (TS). PMID: 23903781
  26. Data suggest that methylenetetrahydrofolate reductase (NAD(P)H), dihydrofolate reductase, thymidylate synthetase, and SLC19A1 genes exhibit increased expression after the highest dose of methotrexate in a laryngeal cancer cell line. PMID: 23838799
  27. Despite structural similarity, Escherichia coli and human DHFRs utilize different dynamic mechanisms to perform the same function, and human DHFR cannot complement DHFR-deficient E. coli cells. PMID: 24077226
  28. Interactions between ligands and Asn 64, Phe 31, and Phe 34 are essential for increased affinity for DHFR. PMID: 24053334
  29. Low activity of endothelial DHFR is a significant factor limiting the benefits of BH4 therapies, which may be further exacerbated by folate supplements. PMID: 23707606
  30. There is an association between DHFR DD/SHMT TT and DHFR II/SHMT TT combined genotypes and folate and MMA concentrations in individuals with Down syndrome. PMID: 23421317
  31. High DHFR immunoexpression correlated with nodal status and primary nasopharyngeal carcinoma. PMID: 23726796
  32. The data presented here provide insights into the evolutionary trajectory of functional DHFR through its protein sequence space, leading to the diverged binding and catalytic properties of the E. coli and human enzymes. PMID: 23733948
  33. Inhibits cell growth through a mechanism involving downregulation of DHFR protein. PMID: 22954684
  34. Genotyping of DHFR 829C>T and GGH -401C>T was performed using a polymerase chain reaction. PMID: 22994778
  35. Leads to accelerated degradation of DHFR and inhibition of cancer cell growth. PMID: 23197646
  36. This study aimed to investigate the potential association between the 19-base pair (bp) deletion polymorphism of the DHFR gene (rs70991108), null genotype of UGT2B17, and the expression level of NGX6 with the risk of breast cancer. PMID: 23053953
  37. Rheumatoid arthritis patients with the DHFR-317AA genotype exhibited a less favorable response to methotrexate. PMID: 22324981
  38. Human dihydrofolate reductase is bound to NADP. PMID: 22024482
  39. Components of the folate cycle may be involved in the etiology of idiopathic intellectual disability. PMID: 22005284
  40. This report clearly demonstrates that the population rate of change of resistant dhfr and dhps alleles is contingent upon sulfadoxine-pyrimethamine usage in the population within the Morogoro-Mvomero district. PMID: 21857842
  41. The 19-bp deletion polymorphism of the DHFR gene was not identified as a maternal risk factor for Down syndrome and was not related to variations in serum folate and plasma homocysteine and methylmalonic acid concentrations in the study population. PMID: 21120433
  42. The 19-base pair deletion polymorphism of DHFR was investigated in a Japanese population. The genotype distribution was as follows: wild/wild, 11.9%; wild/deletion, 40.1%; deletion/deletion, 48.0%. Frequencies of the wild type and deletion alleles were 0.32 and 0.68, respectively. PMID: 20834190
  43. Dihydrofolate reductase deficiency is associated with an inborn error of metabolism. PMID: 21310276
  44. Dihydrofolate reductase deficiency due to a homozygous DHFR mutation causes megaloblastic anemia and cerebral folate deficiency, leading to severe neurologic disease. PMID: 21310277
  45. The first kinetic parameters for DHFR from pjDHFR and pcDHFR with methotrexate (MTX), trimethoprim (TMP), and its potent analogue, PY957, are reported. PMID: 19196009
  46. DNA variants play a role in predisposition to disease-free survival in childhood acute lymphoblastic leukemia. PMID: 19861437
  47. Protein folding of dihydrofolate reductases (DHFR) from human, Escherichia coli, and Lactobacillus casei was elucidated and compared using intrinsic Trp fluorescence and fluorescence-detected ANS binding. PMID: 11779239
  48. This study explored differences between the regulation of Plasmodium and human dihydrofolate reductases. PMID: 11964483
  49. Computer modeling studies investigated the structural role of NADPH binding to active site mutants of human dihydrofolate reductase in complex with piritrexim. PMID: 11996001
  50. This review focuses on molecular mechanisms that govern the translational regulation of DHFR in response to MTX. PMID: 12084458

Show More

Hide All

Database Links

HGNC: 2861

OMIM: 126060

KEGG: hsa:1719

STRING: 9606.ENSP00000396308

UniGene: Hs.592364

Involvement In Disease
Megaloblastic anemia due to dihydrofolate reductase deficiency (DHFRD)
Protein Families
Dihydrofolate reductase family
Subcellular Location
Mitochondrion. Cytoplasm.
Tissue Specificity
Widely expressed in fetal and adult tissues, including throughout the fetal and adult brains and whole blood. Expression is higher in the adult brain than in the fetal brain.

Q&A

What is DHFR and why is it an important research target?

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 .

What are the structural and functional characteristics of human DHFR?

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 .

What are the different types of DHFR antibodies available for research?

Research-grade DHFR antibodies are available in several formats:

Antibody TypeHost SpeciesApplicationsNotable Features
MonoclonalRabbit, MouseWB, IHC, IP, ICC/IFHigher specificity, consistent lot-to-lot performance
PolyclonalRabbit, SheepWB, IHC-P, ICC/IF, IPBroader epitope recognition, potentially higher sensitivity
ConjugatedRabbitWB, ICC/IF, Flow CytometryDirectly labeled with fluorophores like Alexa Fluor® 750

Different antibodies target specific regions of DHFR, including the N-terminus (amino acids 1-50) or regions surrounding particular residues such as Gly175 .

How should DHFR antibodies be optimized for Western blotting applications?

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) .

What are the optimal conditions for DHFR immunohistochemistry and immunocytochemistry?

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

  • Antibody concentration: 5-10 μg/ml for cellular staining

  • Incubation time: 3 hours at room temperature

  • Visualization: Use appropriate fluorophore-conjugated secondary antibodies (e.g., DyLight®550, NorthernLights™ 557)

  • Counterstaining: DAPI for nuclear visualization

  • Validated cell lines: U20S, MCF-7, PC-3 cells show reliable DHFR staining patterns

How can DHFR target engagement be measured in cells?

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:

    • Biochemical assays evaluate direct inhibitory activity

    • Cell-based assays demonstrate target engagement in the cellular environment

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 .

How should researchers validate the specificity of DHFR antibodies?

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:

    • Positive controls: Use recombinant DHFR protein

    • Tissue-specific expression verification: Compare expression levels in tissues known to express DHFR (liver, kidney, colorectal tissue)

    • Cell line panels: Test across multiple cell lines (Jurkat, U937, C6, RAW 264.7, PC-12)

  • 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 .

What are common pitfalls in DHFR antibody-based experiments and how can they be avoided?

Common challenges and solutions include:

  • Non-specific binding:

    • Problem: Background staining in IHC/IF or multiple bands in WB

    • Solution: Optimize blocking (5% NFDM/TBST for WB, 10% goat serum for IHC/IF) and increase antibody dilution

  • Epitope masking:

    • Problem: Reduced or absent signal due to protein-protein interactions

    • Solution: Use multiple antibodies targeting different epitopes; some target the N-terminus (aa 1-50) while others target regions near Gly175

  • Variable expression levels:

    • Problem: DHFR expression can vary across tissues and cell types

    • Solution: Include positive control samples with known DHFR expression (Jurkat, HEK293T)

  • Species cross-reactivity issues:

    • Problem: Unexpected cross-reactivity or lack of expected cross-reactivity

    • Solution: Choose antibodies validated across species of interest; human DHFR shares 90% sequence identity with mouse and rat DHFR

  • Signal detection sensitivity:

    • Problem: Weak signal despite adequate protein expression

    • Solution: Use signal enhancement systems; for IHC, employ HRP Conjugated Super Vision Assay Kit with DAB as chromogen

How can DHFR antibodies be used to study drug resistance mechanisms?

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

What are the latest approaches for using DHFR antibodies in cancer research?

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:

    • Computational modeling identifies promising inhibitor candidates

    • NCI-60 Human Tumor Cell Line Screening data provides efficacy information

    • DHFR antibodies confirm target engagement in cellular contexts

  • 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

How can DHFR antibodies be integrated with other molecular biology techniques for comprehensive studies?

Integrative approaches include:

  • CRISPR/Cas9 gene editing combined with antibody-based detection:

    • Generate DHFR knockout or point mutation cell lines

    • Use DHFR antibodies to confirm knockout efficiency or study mutant protein expression

    • Assess functional consequences on folate metabolism and cell growth

  • Proteomics and interactome studies:

    • Use DHFR antibodies for immunoprecipitation followed by mass spectrometry

    • Identify novel DHFR-interacting proteins under different conditions

    • Map the dynamic DHFR interactome in response to drug treatment

  • Patient-derived xenograft (PDX) models:

    • Use DHFR antibodies to characterize DHFR expression in PDX tumors

    • Correlate expression with drug response

    • Develop personalized treatment strategies based on DHFR status

  • Single-cell analysis techniques:

    • Combine flow cytometry with DHFR antibodies to quantify heterogeneity

    • Link DHFR expression levels to cell cycle phases or differentiation states

    • Identify resistant subpopulations within tumors

  • Computational biology integration:

    • Use antibody-derived expression data to validate in silico predictions

    • Incorporate DHFR expression data into systems biology models

    • Predict synergistic drug combinations targeting DHFR and complementary pathways

How are DHFR antibodies being used to study the relationship between DHFR and microRNAs?

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

What methodological considerations are important when using DHFR antibodies for flow cytometry?

For optimal flow cytometry results with DHFR antibodies:

  • Cell preparation:

    • Fix cells with 4% paraformaldehyde to maintain cellular integrity

    • Permeabilize with appropriate buffer to allow antibody access to intracellular DHFR

  • Blocking conditions:

    • Block with 10% normal goat serum to reduce non-specific binding

    • Use species-matched serum to the secondary antibody host

  • Antibody concentration and incubation:

    • Use 1 μg/1×10⁶ cells for primary antibody

    • Incubate for 30 minutes at 20°C for optimal binding

  • Controls:

    • Include isotype control antibody (e.g., rabbit IgG at 1 μg/1×10⁶ cells)

    • Use unlabelled sample without primary and secondary antibody as blank control

    • Include positive control cell lines with known DHFR expression (PC-3, Jurkat)

  • Secondary antibody selection:

    • Use fluorophore-conjugated secondary antibodies (e.g., DyLight®488)

    • Maintain consistent 5-10 μg/1×10⁶ cells concentration

    • Incubate for 30 minutes at 20°C

  • Data analysis considerations:

    • Analyze shifts in fluorescence intensity to quantify DHFR expression

    • Compare with standard cell lines to normalize expression levels

    • Account for autofluorescence when selecting fluorophores

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.