DHFR (dihydrofolate reductase)

2015-12-01   Maja Krajinovic , Rachid Abaji , Bahram Sharif-Askari 

Identity

HGNC
LOCATION
5q14.1
LOCUSID
ALIAS
DHFRP1,DYR
FUSION GENES

Abstract

Dihydrofolate reductase (DHFR) is a member of the reductase enzyme family, which is ubiquitously expressed in all organisms. Levels of this enzyme peak at the G1\/S cell cycle boundary. Autoregulation, through DHFR-RNA interactions, has also been reported. DHFR catalyzes the NADPH dependent reduction of dihydrofolate (DHF) to tetrahydrofolate (THF) needed for several one-carbon transfer reactions in purine and pyrimidine synthesis (Jensen et al 1997, Klon et al 2002). It is also the only enzyme that reduces folic acid, a synthetic vitamin not found in nature, to dihydrofolate (Banka et al. 2011). Reduction of DHFR enzymatic activity diminishes the THF pool inside the cell which slows DNA synthesis and cell proliferation eventually leading to cell death (Assaraf et al 2007, Klon et al 2002, Morales et al 2009). DHFR inhibition is essential to the action of antifolate medications used to treat cancer and some inflammatory diseases. Changes in DHFR expression can affect susceptibility to a variety of diseases dependent on folate status such as spina bifida and cancer. Likewise, human DHFR (hDHFR) has become a major drug target in anticancer therapy (Klon et al 2002, Sharif-Askari et al 2010).

DNA/RNA

Description

DHFR encoded on chromosome 5 and has 6 exons which are separated by 5 introns.
Chen et al (1984) determined that the DHFR gene is about 30 kb long and consists of 6 exons separated by 5 introns. The full length transcript is 28756 bp long and length of 3-UTR of some human DHFR mRNA molecules was found to be 2900 nucleotides (Chen et al 1984).

Transcription

The human DHFR contains two promoters, the minor transcript which represents only 1% of DHFR mRNA molecules and the major transcript which codes for 99% DHFR mRNA. Independent regulation, low prevalence, nuclear enrichment and low translational efficiency suggest that the DHFR minor transcript may function in vivo to regulate the transcriptional activity of the major promoter (Blume et al 2003 and Martianov et al 2007).

Pseudogene

The DHFR gene family includes the functional DHFR gene and four other intronless pseudogenes, dihydrofolate reductase pseudogene (DHFRP1-4), based on human-rodent somatic cell hybridization. Pseudogene-4 (DHFRP4) is assigned to chromosome 3, pseudogene-1 to chromosome 18 with two transcripts and pseudogene-2 (DHFRP2) to chromosome 6 with one transcript (Anagnou et al. 1984 and Anagnou et al. 1985). Interestingly, according to Anagnou et al. 1988 report, pseudogene-1 (DHFRP1) was found to be present in some individuals while completely absent in others with an interethnic variation in frequency which might implicate a recent origin in the evolutionary process (Anagnou et al., 1988).
Recent studies suggest that DHFRP4, now known as dihydrofolate reductase-like 1 (DHFRL1) is expressed giving a functional protein product which shows a similar but less specific activity to that of DHFR enzyme (McEntee et al 2011).

Proteins

Description

Sequence length: 187 AA. Craik et al. (1983) in DHFR protein study reported that altered surface structures can account for functional differences among the members of a family. Also they pointed out that sliding of the intron-exon junctions may design a mechanism for generating length polymorphisms and divergent sequences.

Expression

DHFR is extensively expressed in the fetal and adult tissues such as heart, liver, skeletal muscle, thymus, kidney, brains and whole blood with higher expression in adult brain in compare to fetal brain (Banka et al 2011).

Localisation

While the dihydrofolate reductase enzyme is thought to be present in multiple cellular compartments, it is most particularly localized in the cytosol and the nucleoplasm.

Function

DHFR is a key enzyme in folate metabolism as it is involved in 5,10-methylene tetra hydro folate (THF) generation from 7,8-dihydrofolate (DHF). The generated 5,10-methylene THF is used for the conversion of deoxyuridylate (dUMP) to deoxythymidylate (dTMP) in a reaction catalyzed by thymidylate synthase (TS). The regenerated THF starts subsequent rounds of thymidylate biosynthesis. Moreover DHFR contributes to the de novo mitochondrial thymidylate biosynthesis pathway and catalyzes de novo glycine and purine synthesis as well as DNA precursor synthesis (Anderson et al 2011 and Assaraf et al 2007).

Homology

Funanage et al (1984) assigned the DHFR gene to chromosome 5 and further narrowed the assignment to 5q11-q22. Based on their evidences, there is a homology between both the short and long arm of hamster chromosome 2 and human chromosome 5. Also, the sequences of the human and mouse DHFR proteins studied by Chen et al was shown to differ in only 21 of 186 amino acids, which reflects an 89% homology in the DNA coding sequences of the genes (Funanage et al 1984 and Chen et al 1984).

Mutations

Germinal

  • Germline polymorphisms
    1. Location: Intron 1
    Polymorphism: 19-bp insertion /deletion (rs70991108
    Impact: Low-serum folate/ high homocysteine, change in mRNA levels
    Related disorders: Neural tube defects and breast cancer
    Note: 19 base pair deletion in intron 1 which elevated risk of developing breast cancer, neural-tube defects (NTD) and may be a risk factor for low birth weight and preterm delivery (Xu et al 2007, Vander linden 2006 and Johnson et al 2005). In contrast, Parle-McDermott et al (2007), demonstrated that 19-bp deletion allele (D) may be a protective genetic factor against NTD by increasing DHFR mRNA levels in pregnant women. Moreover, a study by Rafighdoost also suggests that DHFR 19-bp D/D genotype reduce the risk of Nonsyndromic cleft lip with or without cleft palate (NS-CL/P) in Iranian subjects (Rafighdoost et al. 2015).
    Ongaro et al (2009) and Vagace et al (2011) reported that homozygosity for DHFR 19 bp deleted allele polymorphism has been associated with increased hepatotoxicity in leukemia patients treated with MTX.

    2. Location: 3-UTR
    Polymorphism: C829T, A721T, A1171T
    Impact: MTX resistance
    Related disorder: NTD and Rheumatoid Arthritis
    Note: C829T: Goto et al., (2001), by analyzing 3 untranslated region (UTR) of the human DHFR gene transcript discovered C829T substitution located 223 base pairs downstream from the stop codon and positioned between the first and second polyadenylation site. It interferes with miR-24 function leading to higher DHFR mRNA and protein levels. Present in 14.2% of the Japanese populations, the 829T/T mRNA expression level was found to be higher than 829C/C due to the higher stability of the T/T mRNA (Goto et al, 2001).
    A721T did not have and significant association with NTD, but was found to be in complete linkage disequilibrium (LD) with the 19-bp indel polymorphism. (Parle-McDermott et al 2007).
    Sharma et al (2009) demonstrated that DHFR A1171T (rs7387) polymorphism located in 3UTR is considered as putative predictor for MTX response in rheumatoid arthritis patients.

    3. Location: Downstream to 3UTR
    Polymorphism: A35289G (rs1232027)
    Related disorder: MTX efficacy in patients with psoriatic arthritis.
    Note: Chandran et al (2010) found an association of the A allele of A35289G polymorphism with MTX efficacy in patients with psoriatic arthritis.

    4. Location: Minor promoter
    Polymorphism: C-1610G or T (rs1650694) and A-317/G (rs408626)
    Impact: Higher DHFR expression
    Related disorder: Higher risk of relapse in ALL
    Note: Three polymorphisms in DHFR promoter in the 2 kb region upstream of the first or minor transcription of DHFR gene, (C-1610G/T, C-680A, and A-317G) were found associated with treatment responses in children with acute lymphoblastic leukemia (ALL). Haplotype 1 contains both the A-317 and C-1610 alleles and conferred higher transcriptional activity, as shown by reporter gene assay and quantitative mRNA analysis, likely explaining a worse prognosis in patients carrying this haplotype. The ALL patients who were carriers of this haplotype had reduced event free survival (EFS) (Dulucq et al 2008).

    5. Location: Major promoter
    Polymorphism: G308A (rs1105525), C35T (rs1650697), Length polymorphism 63/91: 9-bp insertion deletion/ 9-bp repeat (rs3045983/ - )
    Impact: Higher DHFR expression
    Related disorder: Higher risk of relapse in ALL
    Note: Six polymorphisms including five SNPs, C35T, C304T, G308A, G319A, and A413G substitutions, along with one length polymorphism composed of two sequence motifs (i.e. insertion/deletion at position 63 and variable number of 9-bp elements at position 91), were identified in the major promoter of DHFR which participate in regulation as both a major promoter and a noncoding minor transcript. Haplotype 1b was identified as a haplotype responsible for the lower relapse-free survival observed in ALL patients. This haplotype is defined by C-1610, C-680, A-317 in the minor promoter and three alleles (T35, A308 and compound length polymorphisms composed of 9-base pair (bp) insertion at position 63 and triple 9bp element at position 91) in the major promoter (Dulucq et al 2008 and Al-Shakfa, et al 2009).

    6. Location: Intron 3
    Polymorphism: A10372C (rs1677693) and A8890G (rs1643659)
    Related disorder: Colorectal cancer (Levine AJ et al 2010).

    Polymorphism: 79940143T>C (rs1643650)
    Related disorder: Rheumatoid Arthritis
    Note: According to Salazar et al, this polymorphism was significantly associated with response to MTX in rheumatoid arthritis patients; patients with C/C and C/T genotypes showed a better response to treatment that those with T/T (Salazar et al 2014).

  • Mutations
    Location: 458A>T (Asp153Val)
    Related disorder: Megaloblastic Anemia.
    Note: Cario et al., 2011 reported a homozygous DHFR mutation, 458A>T (Asp153Val) that leads to DHFR deficiency which in turn results in a complex hematological and neurological disease that can be successfully resolved with folic acid or folinic acid replacement (Banka et al 2011 and Cario et al., 2011).
  • Implicated in

    Entity name
    Megaloblastic anemia
    Note
    Dihydrofolate reductase deficiency is an autosomal recessive metabolic disorder characterized by the hematologic findings of megaloblastic anemia and variable neurologic symptoms.
    A germline missense mutation in DHFR was identified causing subsequent extensive enzyme deficiency and resulting in an inborn error of metabolism which is characterized by megaloblastic anemia and/or pancytopenia, severe cerebral folate deficiency, and cerebral tetrahydrobiopterin deficiency (Banka et al 2011).
    Entity name
    Neural tube defects (NTD)
    Note
    Neural tube closure occurs during a period of rapid cellular proliferation and DHFR activity may be a crucial factor in maintaining optimal DNA synthesis during this time. Changes in the activity of the folate cycle enzymes may affect the folate levels and affect NTD development. The most extensively studied DHFR polymorphism is a 19bp insertion to deletion in the first intron and two polymorphisms within the 3 untranslated region (721A>T and 829C>T) of the DHFR gene (Parle-McDermott et al 2007).
    Entity name
    Rheumatoid Arthritis (RA)
    Note
    Response to treatment with Methotrexate in RA treatment was found to be influenced by the genotypes of the DHFR polymorphisms rs7387 (Sharma et al 2009) and rs1643650 (Salazar et al 2014).
    Entity name
    Breast cancer
    Note
    The DHFR 19-bp deletion polymorphism affects the transcription of DHFR gene in humans which can modify the risk of breast cancer in multivitamin supplement users. A multivitamin supplement has adverse effects in patients carrying the 19-bp insertion allele (Xu et al 2007).
    Entity name
    Colorectal cancer (CRC)
    Note
    Levine et al, 2010 demonstrated significant associations between two DHFR tagSNPs (rs1677693 and rs1643659, located on the third intron of the gene) and CRC risk only in individuals not using multivitamin supplements.
    Entity name
    Retinoblastoma
    Note
    Risk of retinoblastoma was significantly elevated among children of mothers homozygous for the 19bp deletion allele taking prenatal synthetic folic acid supplements (Orjuela, et al 2012).
    Entity name
    Nasopharyngeal carcinoma (NPC)
    Note
    DHFR has a significantly higher expression in NPC and is involved in NPC progression through the nucleotide biosynthetic process (Lee et al 2013).
    Entity name
    Acute lymphoblastic leukemia (ALL)
    Note
    MTX is an important component of maintenance therapy in ALL, it exerts its cytotoxicity function by depletion of reduced folates due to interfering with folate metabolism. Changes in DHFR expression level has been found to correlate with MTX efficacy in ALL (Ongaro et al 2009). Polymorphisms in DHFR gene may affect therapeutic responses to antifolates, leading to lower treatment efficacy or higher adverse drug event frequency. Particular haplotype (1b) increases mRNA levels of DHFR and was associated with a higher risk of ALL relapse.

    Bibliography

    Pubmed IDLast YearTitleAuthors
    198614372009DNA variants in region for noncoding interfering transcript of dihydrofolate reductase gene and outcome in childhood acute lymphoblastic leukemia.Al-Shakfa F et al
    28858111987A novel form of human polymorphism involving the hDHFR-psi 1 pseudogene identifies three RFLPs.Anagnou NP et al
    218761882011Identification of a de novo thymidylate biosynthesis pathway in mammalian mitochondria.Anderson DD et al
    216294352010Dihydrofolate reductase gene variations in susceptibility to disease and treatment outcomes.Askari BS et al
    173333442007Molecular basis of antifolate resistance.Assaraf YG et al
    213102762011Identification and characterization of an inborn error of metabolism caused by dihydrofolate reductase deficiency.Banka S et al
    124617862003The 5'-untranslated RNA of the human dhfr minor transcript alters transcription pre-initiation complex assembly at the major (core) promoter.Blume SW et al
    213102772011Dihydrofolate reductase deficiency due to a homozygous DHFR mutation causes megaloblastic anemia and cerebral folate deficiency leading to severe neurologic disease.Cario H et al
    204729292010Folate pathway enzyme gene polymorphisms and the efficacy and toxicity of methotrexate in psoriatic arthritis.Chandran V et al
    63234481984The functional human dihydrofolate reductase gene.Chen MJ et al
    63442141983Splice junctions: association with variation in protein structure.Craik CS et al
    65040411984Assignment of the human dihydrofolate reductase gene to the q11----q22 region of chromosome 5.Funanage VL et al
    114489092001A novel single-nucleotide polymorphism in the 3'-untranslated region of the human dihydrofolate reductase gene with enhanced expression.Goto Y et al
    93288361997Distinct roles for Sp1 and E2F sites in the growth/cell cycle regulation of the DHFR promoter.Jensen DE et al
    157558372005Common dihydrofolate reductase 19-base pair deletion allele: a novel risk factor for preterm delivery.Johnson WG et al
    120969172002Atomic structures of human dihydrofolate reductase complexed with NADPH and two lipophilic antifolates at 1.09 a and 1.05 a resolution.Klon AE et al
    237267962013Overexpression of thymidylate synthetase confers an independent prognostic indicator in nasopharyngeal carcinoma.Lee SW et al
    206158902010A candidate gene study of folate-associated one carbon metabolism genes and colorectal cancer risk.Levine AJ et al
    172377632007Repression of the human dihydrofolate reductase gene by a non-coding interfering transcript.Martianov I et al
    218761842011The former annotated human pseudogene dihydrofolate reductase-like 1 (DHFRL1) is expressed and functional.McEntee G et al
    191901172009Dihydrofolate reductase amplification and sensitization to methotrexate of methotrexate-resistant colon cancer cells.Morales C et al
    196481632009Gene polymorphisms in folate metabolizing enzymes in adult acute lymphoblastic leukemia: effects on methotrexate-related toxicity and survival.Ongaro A et al
    226489682012Risk of retinoblastoma is associated with a maternal polymorphism in dihydrofolatereductase (DHFR) and prenatal folic acid intake.Orjuela MA et al
    174865952007The 19-bp deletion polymorphism in intron-1 of dihydrofolate reductase (DHFR) may decrease rather than increase risk for spina bifida in the Irish population.Parle-McDermott A et al
    262219212015The 19-bp deletion polymorphism of dihydrofolate reductase (DHFR) and nonsyndromic cleft lip with or without cleft palate: evidence for a protective role.Rafighdoost F et al
    250842012014Polymorphisms in genes involved in the mechanism of action of methotrexate: are they associated with outcome in rheumatoid arthritis patients?Salazar J et al
    199025622009Purine biosynthetic pathway genes and methotrexate response in rheumatoid arthritis patients among north Indians.Sharma S et al
    210641362011Methotrexate-induced subacute neurotoxicity in a child with acute lymphoblastic leukemia carrying genetic polymorphisms related to folate homeostasis.Vagace JM et al
    174131112007A functional 19-base pair deletion polymorphism of dihydrofolate reductase (DHFR) and risk of breast cancer in multivitamin users.Xu X et al
    166720822006Genetic variation in genes of folate metabolism and neural-tube defect risk.van der Linden IJ et al

    Other Information

    Locus ID:

    NCBI: 1719
    MIM: 126060
    HGNC: 2861
    Ensembl: ENSG00000228716

    Variants:

    dbSNP: 1719
    ClinVar: 1719
    TCGA: ENSG00000228716
    COSMIC: DHFR

    RNA/Proteins

    Gene IDTranscript IDUniprot
    ENSG00000228716ENST00000439211P00374
    ENSG00000228716ENST00000439211B0YJ76
    ENSG00000228716ENST00000504396P00374
    ENSG00000228716ENST00000505337P00374
    ENSG00000228716ENST00000505337B0YJ76
    ENSG00000228716ENST00000511032B4DM58

    Expression (GTEx)

    0
    5
    10
    15
    20
    25
    30
    35

    Pathways

    PathwaySourceExternal ID
    One carbon pool by folateKEGGko00670
    Folate biosynthesisKEGGko00790
    One carbon pool by folateKEGGhsa00670
    Folate biosynthesisKEGGhsa00790
    Metabolic pathwaysKEGGhsa01100
    Cell CycleREACTOMER-HSA-1640170
    Cell Cycle, MitoticREACTOMER-HSA-69278
    Mitotic G1-G1/S phasesREACTOMER-HSA-453279
    G1/S TransitionREACTOMER-HSA-69206
    G1/S-Specific TranscriptionREACTOMER-HSA-69205
    E2F mediated regulation of DNA replicationREACTOMER-HSA-113510
    MetabolismREACTOMER-HSA-1430728
    Metabolism of nitric oxideREACTOMER-HSA-202131
    eNOS activation and regulationREACTOMER-HSA-203765
    Tetrahydrobiopterin (BH4) synthesis, recycling, salvage and regulationREACTOMER-HSA-1474151
    Metabolism of vitamins and cofactorsREACTOMER-HSA-196854
    Metabolism of water-soluble vitamins and cofactorsREACTOMER-HSA-196849
    Metabolism of folate and pterinesREACTOMER-HSA-196757
    Antifolate resistanceKEGGko01523
    Antifolate resistanceKEGGhsa01523

    Protein levels (Protein atlas)

    Not detected
    Low
    Medium
    High

    PharmGKB

    Entity IDNameTypeEvidenceAssociationPKPDPMIDs
    PA10810pemetrexedChemicalClinicalAnnotationassociatedPD23709418, 24732178
    PA443434Arthritis, RheumatoidDiseaseClinicalAnnotation, VariantAnnotationassociatedPD19902562, 25084201
    PA443622Carcinoma, Non-Small-Cell LungDiseaseClinicalAnnotationassociatedPD23709418, 24732178
    PA444937MesotheliomaDiseaseClinicalAnnotationassociatedPD24732178
    PA445601OsteosarcomaDiseaseClinicalAnnotationassociatedPD25778468
    PA446155Precursor Cell Lymphoblastic Leukemia-LymphomaDiseaseClinicalAnnotationassociatedPD19648163, 19861437, 21747412, 26335211
    PA450428methotrexateChemicalClinicalAnnotation, VariantAnnotationassociatedPD19648163, 19861437, 19902562, 21747412, 25084201, 25778468, 26335211

    References

    Pubmed IDYearTitleCitations
    172377632007Repression of the human dihydrofolate reductase gene by a non-coding interfering transcript.282
    199131212009Gene-centric association signals for lipids and apolipoproteins identified via the HumanCVD BeadChip.85
    198984822009Genetic variants in TPMT and COMT are associated with hearing loss in children receiving cisplatin chemotherapy.84
    197063812009The extremely slow and variable activity of dihydrofolate reductase in human liver and its implications for high folic acid intake.66
    167354742006Understanding bistability in complex enzyme-driven reaction networks.62
    194933492009118 SNPs of folate-related genes and risks of spina bifida and conotruncal heart defects.62
    120844582002Novel aspects of resistance to drugs targeted to dihydrofolate reductase and thymidylate synthase.53
    119644832002Divergent regulation of dihydrofolate reductase between malaria parasite and human host.49
    126120702003RNA polymerase II accumulation in the promoter-proximal region of the dihydrofolate reductase and gamma-actin genes.47
    174499062007Genetic polymorphisms in one-carbon metabolism: associations with CpG island methylator phenotype (CIMP) in colon cancer and the modifying effects of diet.46

    Citation

    Maja Krajinovic ; Rachid Abaji ; Bahram Sharif-Askari

    DHFR (dihydrofolate reductase)

    Atlas Genet Cytogenet Oncol Haematol. 2015-12-01

    Online version: http://atlasgeneticsoncology.org/gene/40303/dhfr