Molecular genetic analysis of the human dihydrofolate reductase gene: relation with plasma total hom*ocysteine, serum and red blood cell folate levels (2024)

Introduction

Folate, a water-soluble B-vitamin, has gained increasing interest for its essential role in DNA synthesis/repair and the transmethylation pathway. The primary function of folate is as a carrier of one-carbon units for purine and thymidine synthesis and the conversion of hom*ocysteine to methionine. Because of the close interrelation between folate and hom*ocysteine, low folate status is reflected by elevated plasma total hom*ocysteine (tHcy) concentrations.1, 2 A number of diseases have been linked to disturbances in the above-mentioned pathways. For example, low folate status increases the risk of certain cancers3, 4, 5 and is related to congenital malformations, such as neural tube defects.6 Moreover, an increase in tHcy increases the risk of cardiovascular disease and congenital abnormalities.7, 8, 9

Dietary folates, present in reduced and partially reduced form,10 exist as polyglutamates and require hydrolysis to monoglutamates before absorption in the intestine. After release into the portal circulation, folate (mainly as 5-methyltetrahydrofolate) is distributed throughout the body and is used for hom*ocysteine remethylation, leaving tetrahydrofolate (THF) and methionine. Methionine is used for protein synthesis or converted to S-adenosylmethionine, the principle methyl donor in the human body. THF is converted to methyleneTHF, among others, and used for thymidylate synthesis. The latter generates dihydrofolate (DHF) that requires the action of dihydrofolate reductase (DHFR) to be reduced to THF again. In addition, folic acid present in vitamin pills and fortified foods requires full reduction by DHFR via DHF into THF in order to become metabolic active. This makes DHFR an essential enzyme not only for the initial reduction of dietary folates (and folic acid) but also for the continuous reduction of DHF into THF generated during thymidylate synthesis.11

DHFR (EC 1.5.1.3) is located on chromosome 5q11.2-q13.2 and spans about 30 kb. The gene consists of six exons, yielding an mRNA of about 3.9 kb.11 DHFR enzyme (AC 008434.5) is a cytosolic protein of 21.3 kDa and requires NADP as a cofactor. At least three pseudo genes of DHFR are known, that is, DHFRP1 (chromosome 18), DHFRP2 (chromosome 6) and DHFRP4 (chromosome 3, also denoted DHFRL1),12, 13 which are highly hom*ologous to the corresponding DHFR coding regions. Outside these regions, sequence hom*ology is relatively low.14 DHFR deficiency (OMIM 126060) is sporadically seen and results in severe megaloblastic anemia and mild to moderate mental retardation.15, 16 The antifolate methotrexate (MTX) is well known for its inhibitory effect on DHFR thereby preventing DNA synthesis and, hence, tumor growth.17 In addition, inhibition of DHFR by MTX and the antibiotic trimethoprim were shown to increase tHcy levels by 50%,18, 19 suggesting that DHFR dysfunction owing to genetic variants may affect tHcy as well. Recently, Johnson et al.20 described a 19-bp deletion that may affect gene expression.

In this study, we aimed to identify genetic variation within the DHFR gene by sequencing the entire coding region, including the intron–exon boundaries and DHFR flanking regions. Newly identified variants, and the 19-bp deletion in intron 1,20 were assessed for their effect on tHcy and serum and red blood cell (RBC) folate concentrations in population-based controls.

Materials and methods

Study population

DNA from 20 individuals of Caucasian origin, who had participated in an earlier study,21 were used for DHFR sequence analysis. Functional polymorphisms in the DHFR gene are likely to be reflected in plasma folate and hom*ocysteine levels. The subjects were selected based on these criteria: six subjects had low folate (<10th percentile=7.0 nmol/l) and high hom*ocysteine levels (>90th percentile=15.5 μmol/l), nine subjects had a plasma tHcy above 15.5 μmol/l and five subjects were selected randomly. Subjects with low vitamin B12 (<10th percentile=124.7 pmol/l) or the MTHFR 677TT genotype were excluded. We aimed to identify common genetic variants that may contribute to changes in tHcy or folate at the population level. According to a binomial distribution, the chance of finding an allele with a frequency of 2.5% in one out of 40 alleles is 0.64. In addition, the minimum allele frequency required to find at least one out of 40 alleles (with 95% confidence) is 7.2%. The population used for the association study was recruited via a general practice in The Hague (The Netherlands) and has been described in more detail elsewhere.21 In total, 2812 people, 20–90 years of age, were invited to take part in a health survey on risk factors for cardiovascular disease. From the group that agreed to participate (n=532), the first 500 were included. For the current study, DNA was available of 438 individuals. All participants gave their informed consent.

Sequence analysis of the DHFR gene

Blood samples were drawn from the antecubital vein. DNA was extracted,22 and then stored at 4°C. Genomic DNA sequencing of the entire coding region (GenBank accession number NT_006713, AC008434.5) was performed in order to screen the DHFR gene for variation. We also sequenced 665 bp of the 5′-upstream region, which included the 5′-untranslated region (UTR) of about 500 bp (NM_000791), and 1300 bp downstream of DHFR. Intron-based primers were developed to avoid amplification of pseudo genes and to reveal potential splice site variants. Concerning amplification of both 5′ and 3′ UTRs, primer sets were developed taking into account the hom*ology of the known pseudogenes. In addition, alignment of these primer sets showed no matches with any of the known DHFR pseudogenes. PCR amplifications were performed in a total volume of 50 μl on an iCycler (Biorad, The Netherlands). For details, see Table 1. The reaction mixture contained 200 nmol/l of both forward and reverse primer (Biolegio BV, The Netherlands); 200 μmol/l dNTPs, 0.5 U of recombinant Taq polymerase (both from Invitrogen, The Netherlands); PCR buffer containing 20 mmol/l Tris-HCl buffer (pH 8.4), 50 mmol/l KCl and 5% DMSO; 1.0–4.0 mmol/l MgCl2 and 75 ng DNA. An alternative PCR buffer, containing 50 mmol/l Tris-HCl pH 9.2, 16 mmol/l (NH4)2SO4, 2% DMSO and 0.1 % Tween-20 (Roche Applied Science, Switzerland), was used where necessary (see Table 1). PCR conditions were as follows: initial denaturation of 4 min at 94°C, 35 cycles of 94°C/60 s, 55–65°C/30 s, 72°C/30 s, and a final extension of 7 min at 72°C. The PCR products obtained were analyzed on a 2% agarose gel and subsequently sequenced on an ABI Prism 3730 automated sequencer using the ABI Prism Big Dye Terminator cycle sequence kit according to the instructions of the manufacturer (PE Biosystems, The Netherlands). Newly identified variants were confirmed on both DNA strands by direct sequencing.

Full size table

Genetic analysis of DHFR 9-bp repeat

We identified a 9-bp repeat in the 5′-upstream region of the DHFR gene. We obtained the most frequent alleles (3 × , 6 × and 7 × repeat) by cloning PCR fragments (450 bp) into the pGEM-T vector system and transformation of Escherichia coli JM109 High-efficiency Competent cells (Promega, The Netherlands). After ampicillin selection on LB-plates (IPTG/X-gal screening) and plasmid DNA isolation (Wizard plus SV, miniprep kit, Promega), these alleles were used as positive controls in the genotype analyses. In order to facilitate genotyping, a new primer set was chosen for the analysis (F 5′-GCCCAGTCCCAGACAGAAC-3′ and R 5′- GAGCCGATTCTTCCAGTCTAC-3′). The resulting PCR product was 180 bp (for the 6 × repeat allele), allowing a clear distinction between the alleles. PCR conditions: 4 min at 94°C, 35 cycles of 94°C/60 s, 59°C/30 s and 72°C/30 s, and a final extension of 7 min at 72°C. The reaction mixture (50 μl) contained 200 nmol/l of both forward and reverse primer (Biolegio BV, The Netherlands); 200 μmol/l dNTPs, 0.5 U of recombinant Taq polymerase (both from Invitrogen, The Netherlands); PCR buffer containing 50 mmol/l Tris-HCl (pH 9.2), 16 mmol/l (NH4)2SO4, 2% DMSO and 0.1% Tween-20 (Roche Applied Science, Switzerland); 2.0 mmol/l MgCl2 and 75 ng DNA. The PCR products were analyzed on large 3% agarose gels by running the samples at least 4 h at 125 V. In each PCR, DNA samples with a known (sequence confirmed) genotype were run as positive controls. When PCR products were not in control range or difficult to genotype, they were excised and purified from the agarose gel (Gel extraction kit, Qiagen B.V., The Netherlands) and sequenced to identify the length of the allele. Genotype data were obtained from 417 subjects.

Analysis of DHFR intron 1 19-bp deletion

Screening for the 19-bp deletion in intron 1 of the DHFR gene was essentially as described previously.20 In short, the PCR reaction mix (25 μl) contained 75 ng of genomic DNA, 1 × Ampli Taq gold buffer, 1.5 mmol/l MgCl2, 250 μmol/l dNTPs, 1 μmol/l of each primers (FW1: 5′-CCACGGTCGGGGTACCTGCC-3′, FW2: 5′-ACGGTCGGGGTGGCCGACTC-3′ and RV: 5′-AAAAGGGGAATCCAGTCGG-3′), 1 mol/l betaine and 0.5 U Taq polymerase. PCR conditions were as follows: initial denaturation of 4 min at 94°C, 35 cycles of 94°C/60 s, 58°C/60 s, 72°C/60 s, and a final extension of 7 min at 72°C. The PCR products were analyzed on a 3% agarose gel, stained with ethidium bromide and visualized by UV light. On each gel, DNA samples with a known (sequence confirmed) genotype (no del/no del, no del/del, del/del) were run as positive controls. Genotype data were obtained from 330 subjects.

Biochemical parameters

Blood samples were drawn from the antecubital vein for determination of total plasma hom*ocysteine and folate. EDTA samples for hom*ocysteine and folate measurement were placed on ice immediately and centrifuged at 3500 g for 5 min with minimal delay. The plasma or serum was separated and stored at −20°C. Total plasma hom*ocysteine concentrations were measured by an automated high-performance liquid chromatography method with reverse phase separation and fluorescent detection (Gilson 232-401 sample processor, Spectra Physics 8800 solvent delivery system and LC 304 fluorometer).23 DNA extraction was performed as described previously.22 Serum and RBC folate concentrations were measured with the Dualcount Solid Phase No Boil Radioassay (Diagnostic Product Corporation, Los Angeles, CA, USA).

Statistics

tHcy and folate concentrations were logarithmically transformed before all analyses, and are expressed as a geometric mean with a 95% confidence interval (CI). Differences in metabolite concentrations were determined by linear regression analysis and expressed as changes relative to the wild type. A two-tailed P<0.05 was accepted as statistically significant. For all analyses, SPSS 12.0 for Windows was used.

Results

Human DHFR sequence analysis

We sequenced the coding region, including intron–exon boundaries, and UTRs of the DHFR gene (nucleotide −664 to 26702 relative to the A in the translation initiation codon) (see Table 1 for details). We observed no genetic variation within the coding region, but did identify a 9-bp repeat in the highly GC-rich 5′-upstream region of DHFR. The repeat was located 388 bp upstream of the first nucleotide of the translation initiation codon (−388 bp), and partially overlapped with the 5′ UTR. We observed alleles ranging from 162 bp (ie 3 × repeat) to 216 bp (ie 9 × repeat), of which the determined DNA sequences are shown in Figure 1. The most common genotypes (%) were 3/3 (10.6), 3/6 (30.9), 6/6 (41.0) and 6/7 (9.6). Less frequent genotypes (%) were 3/7 (4.1%), 6/9 (1.4%), 3/5, 3/8, 5/6, 6/8 (all 0.5%), 4/7 and 7/7 (both 0.2%). The general consensus sequence of the repeat was A/CGCTGG/CG/AGG/C. In addition, we found three single-nucleotide polymorphisms in the 5′ UTR, one in intron IV and three in 3′ UTR (see Table 2).

DHFR gene structure and DNA sequence of the 5′-upstream region 9-bp repeat as observed for the different alleles.

Full size image
Full size table

Association between DHFR variants and tHcy and folate

Because the 9-bp repeat is located in the promoter region and the 19-bp deletion is thought to affect gene expression,20 we only screened these potentially most important variants for their effect on tHcy, serum and RBC folate in our study population. As shown in Table 3, the 9-bp repeat was not associated with tHcy in our study population. Although the 3/6 and 3/7 repeat compared to the 3/3 repeat genotype showed a trend towards higher plasma folate levels (P trend=0.015 for the 3/3, 3/6 and 3/7 repeat genotype), this increment did not persist in the 6/6 and 6/7 repeat genotypes. The 19-bp deletion was associated with a 2.5% (95% CI: −10.8 to 6.5) and 14.4% (95% CI: −23.5 to −4.5) decrease in tHcy for the heterozygous and hom*ozygous genotype, respectively, compared with the wild-type genotype (P ANOVA=0.016)(Table 3). Adjustment for age and sex did not change the association between this deletion and tHcy. Serum and RBC folate levels were similar between the genotypes. No interaction between the 9-bp repeat and 19-bp deletion or between low folate (<30th percentile, 10.0 nmol/l) and either variant on tHcy was observed (not shown).

Full size table

Discussion

In this study, we searched for genetic variation in the DHFR gene that may affect tHcy, serum and RBC folate levels. We identified a 9-bp repeat in the 5′-upstream region of DHFR that partially overlapped with the 5′ UTR. The 9-bp repeat may affect mRNA stability or translation efficiency, similarly as described for the 28-bp repeat in the functionally related thymidylate synthase.24 The repeat appeared highly polymorphic with alleles ranging from 3 to 9 repeats. In 1995, Nakajima et al.25 described, in Japanese subjects, a 9-bp repeat in exon 1 of the DNA repair gene mutS hom*olog3, which partially overlaps with the DHFR gene and may share promoter elements. Our repeat most likely represents the same repeat, as the consensus sequence highly resembled that reported by Nakajima, although higher repeat sizes were present in our, predominantly Caucasian, population. The repeat may have arisen from duplications of one (or more) ancestral allele(s) or result from DNA replication errors. Despite its location in the promoter region, the length of this repeat was not associated with tHcy, serum and RBC folate levels.

In 2001, Goto et al.26 reported a functional variant (829C>T) in the 3′ UTR of DHFR. We did not observe this variant in the subjects that were included in the sequence analysis, suggesting that it is a population-specific polymorphism or has a minor allele frequency below 2.5%. Johnson et al.20, 27 identified a 19-bp deletion in the highly conserved intron 1, which may be a maternal risk factor for having a child with spina bifida and preterm delivery. It has been postulated that this variant deletes a putative SP1 transcription factor-binding site (required for transcriptional activation/repression) or may affect the splicing process. We show that the 19-bp del/del genotype is associated with a reduced plasma tHcy of about 1.5 μmol/l (14%) compared to wild-type subjects. This may suggest that the 19-bp deletion indeed increases DHFR expression thereby facilitating hom*ocysteine remethylation. Unfortunately, no RNA was available from our study population to support its effect at the expression level, which warrants further investigation.

We detected several other variants in non-coding regions of DHFR which, based on the sequence, could not be ascribed to known pseudogenes. The main part of the introns has not been screened, but these may contain functional variants as well. It is possible that intronic polymorphisms represent a functional variant, although the intronic sequences, except for intron 1, are not well-conserved across species.28 We assessed linkage disequilibrium (LD) between the 19-bp deletion and the most common alleles of the 9-bp repeat (3 × and 6 ×), and found them to be in almost complete LD (D′ 0.9). However, the (squared) correlation coefficient was low (R2=0.3). The HapMap consortium investigated LD between existing DHFR single-nucleotide olymorphisms (http://www.hapmap.org/), and showed that one haplotype block of about 24 kb almost completely covers the DHFR gene. This suggest that the number of variants of the DHFR gene present in the population is limited and may be captured by genotyping just a small number of polymorphisms in the gene.

In conclusion, the coding region of DHFR harbors no variation in our population, which illustrates its importance in folate metabolism. Several variants were found within the non-coding regions of DHFR, potentially the most interesting being the 9-bp repeat in 5′ UTR. The 19-bp deletion in intron 1 was associated with lower tHcy levels and may be a genetic risk factor for cardiovascular disease and cancer, in addition to its reported effect on neural tube defect risk.

Accession codes

Accessions

GenBank/EMBL/DDBJ

References

  1. Mason JB : Biomarkers of nutrient exposure and status in one-carbon (methyl) metabolism. J Nutr 2003; 133 (Suppl 3): 941S–947S.

    Article CAS Google Scholar

  2. Verhoef P, de Groot LC : Dietary determinants of plasma hom*ocysteine concentrations. Semin Vasc Med 2005; 5: 110–123.

    Article Google Scholar

  3. Blount BC, Mack MM, Wehr CM et al: Folate deficiency causes uracil misincorporation into human DNA and chromosome breakage: implications for cancer and neuronal damage. Proc Natl Acad Sci USA 1997; 94: 3290–3295.

    Article CAS Google Scholar

  4. Duthie SJ, Narayanan S, Sharp L, Little J, Basten G, Powers H : Folate, DNA stability and colo-rectal neoplasia. Proc Nutr Soc 2004; 63: 571–578.

    Article CAS Google Scholar

  5. Friso S, Choi SW : Gene-nutrient interactions in one-carbon metabolism. Curr Drug Metab 2005; 6: 37–46.

    Article CAS Google Scholar

  6. Czeizel AE, Dudas I : Prevention of the first occurrence of neural-tube defects by periconceptional vitamin supplementation. N Engl J Med 1992; 327: 1832–1835.

    Article CAS Google Scholar

  7. Wald DS, Law M, Morris JK : hom*ocysteine and cardiovascular disease: evidence on causality from a meta-analysis. BMJ 2002; 325: 1202.

    Article Google Scholar

  8. den Heijer M, Lewington S, Clarke R : hom*ocysteine, MTHFR and risk of venous thrombosis: a meta-analysis of published epidemiological studies. J Thromb Haemost 2005; 3: 292–299.

    Article CAS Google Scholar

  9. de la Calle M, Usandizaga R, Sancha M, Magdaleno F, Herranz A, Cabrillo E : hom*ocysteine, folic acid and B-group vitamins in obstetrics and gynaecology. Eur J Obstet Gynecol Reprod Biol 2003; 107: 125–134.

    Article CAS Google Scholar

  10. Seyoum E, Selhub J : Properties of food folates determined by stability and susceptibility to intestinal pteroylpolyglutamate hydrolase action. J Nutr 1998; 128: 1956–1960.

    Article CAS Google Scholar

  11. Chen MJ, Shimada T, Moulton AD et al: The functional human dihydrofolate reductase gene. J Biol Chem 1984; 259: 3933–3943.

    CAS Google Scholar

  12. Chen MJ, Shimada T, Moulton AD, Harrison M, Nienhuis AW : Intronless human dihydrofolate reductase genes are derived from processed RNA molecules. Proc Natl Acad Sci USA 1982; 79: 7435–7439.

    Article CAS Google Scholar

  13. Anagnou NP, O'Brien SJ, Shimada T, Nash WG, Chen MJ, Nienhuis AW : Chromosomal organization of the human dihydrofolate reductase genes: dispersion, selective amplification, and a novel form of polymorphism. Proc Natl Acad Sci USA 1984; 81: 5170–5174.

    Article CAS Google Scholar

  14. Yang JK, Masters JN, Attardi G : Human dihydrofolate reductase gene organization. extensive conservation of the G+C-Rich 5′ non-coding sequence and strong intron size divergence from hom*ologous mammalian genes. J Mol Biol 1984; 176: 169–187.

    Article CAS Google Scholar

  15. Tauro GP, Danks DM, Rowe PB et al: Dihydrofolate reductase deficiency causing megaloblastic anemia in two families. N Engl J Med 1976; 294: 466–470.

    Article CAS Google Scholar

  16. Erbe RW : Inborn errors of folate metabolism (second of two parts). N Engl J Med 1975; 293: 807–812.

    Article CAS Google Scholar

  17. Banerjee D, Ercikan-Abali E, Waltham M et al: molecular mechanisms of resistance to antifolates, a review. Acta Biochim Pol 1995; 42: 457–464.

    CAS Google Scholar

  18. Kishi S, Griener J, Cheng C et al: hom*ocysteine, pharmacogenetics, and neurotoxicity in children with leukemia. J Clin Oncol 2003; 21: 3084–3091.

    Article CAS Google Scholar

  19. Smulders YM, de Man AM, Stehouwer CD, Slaats EH : Trimethoprim and fasting plasma hom*ocysteine. Lancet 1998; 352: 1827–1828.

    Article CAS Google Scholar

  20. Johnson WG, Stenroos ES, Spychala JR, Chatkupt S, Ming SX, Buyske S : New 19 bp deletion polymorphism in intron-1 of dihydrofolate reductase (DHFR): a risk factor for spina bifida acting in mothers during pregnancy? Am J Med Genet 2004; 124A: 339–345.

    Article Google Scholar

  21. den Heijer M, Blom HJ, Gerrits WB et al: Is hyperhom*ocysteinaemia a risk factor for recurrent venous thrombosis? Lancet 1995; 345: 882–885.

    Article CAS Google Scholar

  22. Miller SA, Dykes DD, Polesky HF : A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res 1988; 16: 1215.

    Article CAS Google Scholar

  23. te Poele-Pothoff MT, van den Berg M, Franken DG et al: Three different methods for the determination of total hom*ocysteine in plasma. Ann Clin Biochem 1995; 32: 218–220.

    Article CAS Google Scholar

  24. Horie N, Aiba H, Oguro K, Hojo H, Takeishi K : Functional analysis and dna polymorphism of the tandemly repeated sequences in the 5′-terminal regulatory region of the human gene for thymidylate synthase. Cell Struct Funct 1995; 20: 191–197.

    Article CAS Google Scholar

  25. Nakajima E, Orimo H, Ikejima M, Shimada T : Nine-bp repeat polymorphism in exon 1 of the HMSH3 gene. Jpn J Hum Genet 1995; 40: 343–345.

    Article CAS Google Scholar

  26. Goto Y, Yue L, Yokoi A et al: A novel single-nucleotide polymorphism in the 3′-untranslated region of the human dihydrofolate reductase gene with enhanced expression. Clin Cancer Res 2001; 7: 1952–1956.

    CAS Google Scholar

  27. Johnson WG, Scholl TO, Spychala JR, Buyske S, Stenroos ES, Chen X : Common dihydrofolate reductase 19-base pair deletion allele: a novel risk factor for preterm delivery. Am J Clin Nutr 2005; 81: 664–668.

    Article CAS Google Scholar

  28. Masters JN, Attardi G : The nucleotide sequence of the CDNA coding for the human dihydrofolic acid reductase. Gene 1983; 21: 59–63.

    Article CAS Google Scholar

Download references

Acknowledgements

This study was supported by Grant 2002B68 from The Netherlands Heart Foundation. Martin den Heijer, MD, PhD, is supported by a VENI-grant from the Dutch Organization for Scientific Research (NWO).

Author information

Authors and Affiliations

  1. Laboratory of Paediatrics and Neurology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands

    Henkjan Gellekink,Henk J Blom&I J M van der Linden

  2. Department of Endocrinology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands

    Henkjan Gellekink&Martin den Heijer

  3. Department of Epidemiology and Biostatistics, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands

    Martin den Heijer

Authors

  1. Henkjan Gellekink

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  2. Henk J Blom

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  3. I J M van der Linden

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  4. Martin den Heijer

    View author publications

    You can also search for this author in PubMedGoogle Scholar

Corresponding author

Correspondence to Martin den Heijer.

Rights and permissions

About this article

Cite this article

Gellekink, H., Blom, H., van der Linden, I. et al. Molecular genetic analysis of the human dihydrofolate reductase gene: relation with plasma total hom*ocysteine, serum and red blood cell folate levels. Eur J Hum Genet 15, 103–109 (2007). https://doi.org/10.1038/sj.ejhg.5201713

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/sj.ejhg.5201713

Keywords

  • folate
  • dihydrofolate reductase
  • genetic variation
  • hom*ocysteine
Molecular genetic analysis of the human dihydrofolate reductase gene: relation with plasma total hom*ocysteine, serum and red blood cell folate levels (2024)

FAQs

What is the function of dihydrofolate reductase in humans? ›

Dihydrofolate reductase (DHFR) is required to maintain the reduced folates necessary for donating carbon atoms, which are essential for the de novo synthesis of purines, pyrimidines, and amino acids, and is the intracellular target of four amino analogs of folic acid (i.e., antifolates).

What is DHFR and why is it important? ›

Dihydrofolate reductase (DHFR) is a key enzyme in folate metabolism and, therefore, in the production of thymidine [2,3]. Its role in thymidine biosynthesis is the reduction of dihydrofolate to tetrahydrofolate using the cofactor NADPH (Fig. 1).

What is dihydrofolate reductase deficiency? ›

Dihydrofolate reductase deficiency (DHFR deficiency) is a rare inherited disorder of folate metabolism caused by defects in the DHFR gene.

What drugs inhibit dihydrofolate reductase? ›

III Examples of Antifolate Drugs that Target Dihydrofolate Reductase
DrugTarget enzyme
MethotrexateDHFR
Edatrexate (10-EDAM)DHFR
MTN (LY231514)TS DHFR GARFT
PT523DHFR
5 more rows

What drugs are acting on DHFR? ›

The two main classical dihydrofolate reductase inhibitors are aminopterin (AM) and methotrexate (MTX, amethopterin), which were designed by replacing lactim-type OH group at C-4 of the natural substrate (DHF) by an amino group.

What enzymes use folate? ›

Folate plays a critical role in DNA synthesis and methylation. Intracellular folate homeostasis is maintained by the enzymes folylpolyglutamate synthase (FPGS) and γ-glutamyl hydrolase (GGH).

What is congenital dihydrofolate deficiency? ›

Dihydrofolate reductase deficiency is an autosomal recessive metabolic disorder characterized by the hematologic findings of megaloblastic anemia and variable neurologic symptoms, ranging from severe developmental delay and generalized seizures in infancy (Banka et al., 2011) to childhood absence epilepsy with learning ...

What is the DHFR gene mutation? ›

Dihydrofolate reductase deficiency due to a hom*ozygous DHFR mutation causes megaloblastic anemia and cerebral folate deficiency leading to severe neurologic disease. Dihydrofolate reductase deficiency is associated with inborn error of metabolism.

What is the most effective compound on DHFR? ›

Quinazoline 94 was found to be the most potent inhibitor of DHFR enzyme (IC50 = 0.4 μM) and produced moderate growth inhibition (GI50 = 23.5 μM) of various cancer cell lines. Similar mode and strength of binding as that of standard drug methotrexate was perceived in the hDHFR pocket in docking studies.

What is reductase deficiency syndrome? ›

5-alpha reductase deficiency is a condition that affects sexual development before birth and during puberty. People with this condition are genetically male, with one X and one Y chromosome in each cell, and they have male gonads (testes) .

Where does DHFR live? ›

In humans, the DHFR enzyme is encoded by the DHFR gene. It is found in the q14. 1 region of chromosome 5.

What are the symptoms of dihydrobiopterin reductase deficiency? ›

Dihydropteridine reductase (DHPR) deficiency (OMIM: 261630) is an autosomal recessive disorder in the regeneration pathway of tetrahydrobiopterin (BH4). Individuals with DHPR deficiency may present with hyperphenylalaninemia, microcephaly, hypotonia, mental retardation, and convulsions (Blau et al.

Do humans have dihydrofolate reductase? ›

Human dihydrofolate reductase (DHFR) was previously thought to be the only enzyme capable of the reduction of dihydrofolate to tetrahydrofolate; an essential reaction necessary to ensure a continuous supply of biologically active folate.

What is a serious side effect of methotrexate? ›

Serious side effects

a persistent cough, chest pain, difficulty breathing, or you become breathless – these may be signs of inflammation of your lungs. swollen hands, ankles or feet, changes to how often you pee or not peeing at all – these may be signs of kidney problems.

What is the activity of dihydrofolate reductase? ›

Dihydrofolate reductase (DHFR) catalyzes the reduction of dihydrofolate to tetrahydrofolate (THF). THF is needed for the action of folate-dependent enzymes and is thus essential for DNA synthesis and methylation.

What is the mechanism of action of dihydrofolate reductase? ›

DHFR catalyzes the reduction of 7,8-dihydrofolate (H2F) to 5,6,7,8-tetrahydrofolate (H4F) through a stereospecific transfer of the pro-R hydrogen from the cofactor nicotinamide adenine dinucleotide phosphate (NADPH; also abbreviated as NH) to the C6 atom of the pterin nucleus, with concomitant protonation at N5 (Figure ...

What is the function of dihydrofolate reductase DHFR )? ›

Dihydrofolate reductase (DHFR) catalyzes the reduction of dihydrofolate to tetrahydrofolate (THF). THF is needed for the action of folate-dependent enzymes and is thus essential for DNA synthesis and methylation.

What is the activity of DHFR? ›

DHFR catalyzes the transfer of a hydride from NADPH to dihydrofolate with an accompanying protonation to produce tetrahydrofolate. In the end, dihydrofolate is reduced to tetrahydrofolate and NADPH is oxidized to NADP+.

How does the DHFR system work? ›

How the DHFR System works. The diagram to the left shows how the DHFR enzyme catalyses the conversion of folate to tetrahydrofolate. This precursor is necessary for de-novo synthesis of purines and pyrimidines (Goeddel, 1990), allowing cells to replicate.

References

Top Articles
Easiest Rustic Bread Recipe EVER | Simple & Delicious! - Shelf Cooking
Keto Egg Roll in a Bowl -- Easy One Pot Recipe
Aberration Surface Entrances
Obor Guide Osrs
Wild Smile Stapleton
craigslist: south coast jobs, apartments, for sale, services, community, and events
Bluegabe Girlfriend
How to Type German letters ä, ö, ü and the ß on your Keyboard
Culver's Flavor Of The Day Monroe
Cincinnati Bearcats roll to 66-13 win over Eastern Kentucky in season-opener
Nier Automata Chapter Select Unlock
Blog:Vyond-styled rants -- List of nicknames (blog edition) (TouhouWonder version)
Notisabelrenu
Inevitable Claymore Wow
Where does insurance expense go in accounting?
Aspen.sprout Forum
O'reilly's Auto Parts Closest To My Location
What Happened To Anna Citron Lansky
N2O4 Lewis Structure & Characteristics (13 Complete Facts)
Kitty Piggy Ssbbw
Las 12 mejores subastas de carros en Los Ángeles, California - Gossip Vehiculos
Zalog Forum
Woodmont Place At Palmer Resident Portal
Ice Dodo Unblocked 76
Craigslist St. Cloud Minnesota
Jermiyah Pryear
Southwest Flight 238
January 8 Jesus Calling
Evil Dead Rise Ending Explained
John Deere 44 Snowblower Parts Manual
John Philip Sousa Foundation
Hannah Jewell
Lincoln Financial Field, section 110, row 4, home of Philadelphia Eagles, Temple Owls, page 1
Loopnet Properties For Sale
What does wym mean?
Exploring TrippleThePotatoes: A Popular Game - Unblocked Hub
Lovein Funeral Obits
Cpmc Mission Bernal Campus & Orthopedic Institute Photos
Stranahan Theater Dress Code
814-747-6702
✨ Flysheet for Alpha Wall Tent, Guy Ropes, D-Ring, Metal Runner & Stakes Included for Hunting, Family Camping & Outdoor Activities (12'x14', PE) — 🛍️ The Retail Market
Wgu Admissions Login
Gt500 Forums
DL381 Delta Air Lines Estado de vuelo Hoy y Historial 2024 | Trip.com
Random Warzone 2 Loadout Generator
Craigslist Marshfield Mo
Washington Craigslist Housing
Assignation en paiement ou injonction de payer ?
Coleman Funeral Home Olive Branch Ms Obituaries
How to Get a Check Stub From Money Network
Latest Posts
Article information

Author: Terence Hammes MD

Last Updated:

Views: 6058

Rating: 4.9 / 5 (69 voted)

Reviews: 84% of readers found this page helpful

Author information

Name: Terence Hammes MD

Birthday: 1992-04-11

Address: Suite 408 9446 Mercy Mews, West Roxie, CT 04904

Phone: +50312511349175

Job: Product Consulting Liaison

Hobby: Jogging, Motor sports, Nordic skating, Jigsaw puzzles, Bird watching, Nordic skating, Sculpting

Introduction: My name is Terence Hammes MD, I am a inexpensive, energetic, jolly, faithful, cheerful, proud, rich person who loves writing and wants to share my knowledge and understanding with you.