SLC19A1 Pharmacogenomics Summary
Folate is a crucial nutrient that supports important physiological functions such as DNA synthesis, cell division and substrate methylation. Low folate level caused by suboptimal intake, transport and cellular utilization of folate, is a risk factor for diseases such as spina bifida and cardiovascular diseases [1]. Intracellular uptake of folate is in part mediated by the reduced folate carrier protein 1 (RFC1), encoded by the human solute carrier family 19, member 1 (SLC19A1) gene. RFC1 is a high-capacity, bi-directional transporter of 5-methyl-tetrahydrofolate and thiamine monophosphate. RFC1 also actively transports antifolate chemotherapeutic agents, such as methotrexate (MTX), into cells [2–9]. In addition to its role in folate uptake, RFC1 plays critical role in folate homeostasis of mammalian cells, where it is down regulated in response to folate deficiency [10].
SLC19A1 is found on chromosome 21 (21q22.3). Several groups of investigators have cloned cDNAs (approximately 2.7 kb in length) encoding the 591-amino acid human SLC19A1 from lymphoblast, placenta, and small intestine cDNA libraries [2–5]. The SLC19A1 gene from human lymphoblasts was shown to contain five exons (exon 2 to exon 6), which code for RFC1 protein [11–13]. There are at least four 5-prime alternative exons, which are used in the production of RFC1 mRNA transcript in lymphoblast cells. These are spliced in a mutually exclusive manner to exon 2, which contains the translational start site for the RFC1 protein. Semi-quantitative PCR shows that exon 1 is preferentially incorporated in the transcript [11]. A separate study showed that RFC1 exhibits alternative splicing [14]. Specifically, three splice variants of RFC1 were identified from a human liver genomic library, and resulted from the incorporation of three alternatives of exon 1 and different 3' sequences. Functional deletion analysis of the region upstream of the transcriptional start site of the SLC19A1 gene led to the identification of two TATA-less promoters, each of which showed significant differences in the efficiency of transcription.
A humanized mouse model for the reduced-function folate carrier has been created [15]. Expressing human SLC19A1 in transport-deficient Chinese hamster ovary cells results in restoration of MTX transport and MTX sensitivity [4]. Several point mutations have been identified in SLC19A1 and down regulation of SLC19A1 mRNA has also been associated with impaired MTX transport and MTX resistance [16–19]. A reverse correlation between the RFC promoter methylation and its mRNA level in cancer cell lines has been described [20]. These constitute important factors in the development of resistance to anti-folate chemotherapeutic agents. A recent study in polymorphism and expressions of folate pathway genes showed that SLC19A1 expression correlated with the sensitivity of several drugs (antifolates, thiopurines, nitrosoureas, and DACH-platinum drugs) in the NCI-60 cancer cell lines [21]. Furthermore, some groups have also found association between the expression of RFC1 and accumulation of methotrexate in Acute Lymphocytic Leukemias (ALL) cells [22, 23]. The SLC19A1 mRNA expression from ALL blasts isolated from newly diagnosed children were higher in certain lineage ALL such as the hyperdiploid B-lineage compared to nonhyperdiploid ALL. In addition, the accumulation of methotrexate polyglutamates was highest in the hyperdiploid B-lineage which indicated that higher SLC19A1 expression plays important role in MTX accumulation [22, 23].
Sequencing of SLC19A1 in healthy subjects
The SLC19A1 gene is highly polymorphic in humans. The proximal promoter region, exons 3 to 5 and their flanking intronic regions of the SLC19A1 gene has been resequenced in an ethnically diverse population of 276 individuals as part of the Pharmacogenetics of Membrane Transporters (PMT) project (http://pharmacogenetics.ucsf.edu/cgi-bin/Study.py). This cohort includes unrelated healthy individuals from the San Francisco Bay Area (80 African Americans, 80 European Americans, 60 Asian Americans, 50 Mexican Americans and 6 Pacific Islanders). A total of 6 non-synonymous SNPs were found in exons 3 to 5 of SLC19A1 gene in this cohort. (https://www.pharmgkb.org/do/serve?objId=PA327&objCls=Gene#tabview=tab2 and in http://pharmacogenetics.ucsf.edu/). Among the non-synonymous SNPs, there are 4 singletons (only found on one chromosome from the sequenced SOPHIE cohort) and they are Leu338Phe (1012C>T, rs59638403, 0.6% in African American); Gly341Asp (1022G>A; rs56822323, 0.6% in European American); Cys458Gly (1372T>G; rs58227024, 0.6% in European American) and Asp522Asn (1564G>A; rs58836581, 0.6% in African American) and two rare variants with minor allele frequency of 1% (Arg456Gln; 1367G>A; rs59841046 and Ala469Val; 1406C>T; rs7278825). One common non-synonymous variant (Ala558Val; 1792C>T; rs35786590) is reported in the dbSNP (dbSNP 130) with total allele frequency of 49.9% and it is found across the four HapMap populations (CEU, HCB, JPT and YRI) (http://www.ncbi.nlm.nih.gov/projects/SNP/snp_ref.cgi?rs=35786590). A frameshift as a result of deletion of 52bp in Exon 3 and give rise to a synonymous SNP (Ala324Ala; 972G>A; rs56138890) is found in the Asian American (12.7 %) and in Mexican American (3 %) populations of the SOPHIE cohort (http://pharmacogenetics.ucsf.edu/) and also reported in the Indian population in India (dbSNP Build 131, http://www.ncbi.nlm.nih.gov/SNP/snp_ref.cgi?rs=56138890). A common non-synonymous SNP in Exon 2 of the gene, Arg27His (80G>A; rs1051266), which was not sequenced in the PMT project, but it is reported in the dbSNP, have total minor allele frequency of 44% and is found across all ethnic groups. In addition, the sequencing of SLC19A1 promoter region in 72 healthy individuals in Australia revealed a 61-bp insertion polymorphisms in 78% of these individuals. Functional studies conducted by this group showed that this 61-bp insertion resulted in higher luciferase activity as a result from the additional binding sites for AP-2 and Mzf-1 transcription factors in this 61-bp insertion region [13].
Sequencing of SLC19A1 in resistant cell lines and tumor specimens
Several SLC19A1 gene mutations leading to antifolate-resistant phenotype are observed in rodent and human cell lines [6, 16, 24]. In one study, L1210 mouse leukemia cells were selected for resistance to the anti-folate (6R)-5,10-dideazatetrahydrofolate [16]. The resistant mouse leukemia cells were shown to have two point mutations in SLC19A1, I48F and W105G, which contributed to the drug-resistant phenotype. Since these mutations dramatically altered the kinetics of folate transport, they were reported as participating in substrate interaction and likely constitute the substrate recognition domain of RFC1. Since the L1210/D3 resistant cells were shown to express the mutant transporter but the wild-type allele was still present in the genomic DNA, it seems that the wild-type allele was silenced during the development of resistance. Another point mutation in SLC19A1 was identified in a methotrexate transport-defective human T-ALL cell line known as CCRF-CEM [24]. This cell line has a lysine to glutamic acid substitution at codon 45 (E45K) and has been identified to be resistant to other anti-folate [16, 25]. Although this nonsynonymous variant could lead to methotrexate resistance due to decreased membrane transport, and have altered binding affinities and transport of folate substrates, there was no evidence that this mutation occurred in ALL samples from children [24]. Other point mutations have been discovered by sequence analysis in human CEM leukemia cells which is resistant to a thymidylate synthase inhibitor (GW1843) and methotrexate. The mutations appeared to be Val29Leu, Glu45Lys and Ser46Ile in the first transmembrane domain of the SLC19A1 and conferred resistance to GW1843 in cells transfected with these mutations [26]. Similarly, other point mutations in SLC19A1 have also been reported in human leukemia cells after cellular exposure to gradually increasing PT523, a potent dihydrofolate reductase inhibitor. DNA sequencing of these cells revealed mutations in SLC19A1: Leu143Pro, Ala147Val, Arg148Gly, Gln150Stop and these mutations resulted in decreased RFC protein levels and showed impaired methotrexate transport [27]. On the other hand, a few studies have been conducted where RFC1 were resequenced in tumor samples. In one study, 203 B-precursor ALL specimens, 32 T-lineage ALL specimens and 11 AML specimens were resequenced for its RCF1. Only 3 B-precursor ALL specimens were found to have nonsynonymous variants, i.e. Asp56His and Asp522Asn [28]. The study showed that the methotrexate uptake is lower in the blast cells from B-precursor ALL patient with the Asp522Asn mutation compared with cells from the wild type carriers. In another study, RFC1 was resequenced in 162 osteosarcoma samples, and the study has identified new variants, such as Ser46Asn, Glu21Lys, Ala7Val and Ser4Pro, in addition to the common variant Arg27His. [29]. In addition, the clinical significance of these genetic polymorphisms in terms of methotrexate plasma level and resistance has not been determined.
Table 1 summarizes the functional effect of genetic variants in SLC19A1 gene identified through sequencing project in healthy subjects, in patients and in antifolate resistant cells.
Table 1.
Summary of SLC19A1 variants in healthy subjects (germline polymorphisms) | |||
---|---|---|---|
SNP | Source | Function/Effect | Allele Frequency |
rs1051266 (Arg27His) | HapMap database and dbSNP (Build 131)c | G allele: 55% (European); 50% (Han Chinese); 43.2% (Japanese); 25.8% (Sub-Saharan African) | |
rs59638403 (Leu338Phe) | PMT sequencing project in unrelated healthy individualsa, b | Not characterized | 0.6% in African American |
rs56822323 (Gly341Asp) | PMT sequencing project in unrelated healthy individualsa, b | Not characterized | 0.6% in African American |
rs58227024 (Cys458Gly) | PMT sequencing project in unrelated healthy individualsa, b | Not characterized | 0.6% in African American |
rs58836581 (Asp522Asn) | PMT sequencing project in unrelated healthy individualsa, b | Not characterized | 0.6% in African American |
rs59841046 (Arg456Gln) | PMT sequencing project in unrelated healthy individualsa, b | Not characterized | 1.7% in Asian American |
rs7278825 (Ala469Val) | PMT sequencing project in unrelated healthy individualsa, b | Not characterized | 1% in Mexican and 0.8% in Asian |
rs56138890 (Ala324Ala) | PMT sequencing project in unrelated healthy individualsa, b and in Indian population in India (dbSNP Build 131)c | Not characterized | 12.7% Asian American; 3% in Mexican American; 14.9% Indian |
61bp insertion in -715 and -714 of SLC19A1 basal promoter (GenBank: AF046920.1) | In 72 healthy subjects | Increase in promoter activity [13]. | 78% with this insertion. The ethnicity of the subjects was not mentioned. |
Summary of SLC19A1 variants identified in tumor specimense | |||
SNP | Source | Function/Effect | Allele Frequency |
Asp56His and Asp522Asn (rs58836581) | 3 B-precursor ALL out of 203 B-precursor ALL, 32 T-lineage ALL specimens and 11 AML specimensd,e | The uptake of methotrexate in blast cells from patients with Asp522Asn variant is significantly lowered compared to other samples containing reference SLC19A1 gene [28]. | Asp167His: 1 patient; Asp522Asn: 1 patient |
Ser46Asn, Glu21Lys, Ala7Val, Ser4Pro and Arg27His (rs1051266) | 162 osteosarcoma samples | The effect of these variants with methotrexate plasma level and resistance cannot be ruled out [29]. | Arg27/Arg27: 30.2%; His27/His27: 22.8%; His27/Arg27: 37.6%; Ser46Asn: 4.32%; Glu21Lys: 1.85%; Ala7Val: 1.23%; Ser4Pro: 1.23%; |
Summary of point mutations of SLC19A1 identified by acquired resistance to various antifolates in sublinese | |||
SNP | Source | Function/Effect | Allele Frequency |
Ile46Phe and Trp105Gly | Point mutations in L1210 mouse leukemia cells. | Transfection of the SLC19A1 cDNA containing these mutants in L1210 cells conferred resistance to 5,10-dideazatetrahydrofolate compound [16, 25]. | Not determined |
Arg27His (rs1051266), Val29Leu, Glu45Lys and Ser46Ile, Leu143Pro, Ala147Val, Arg148Gly, Gln150Stop, Ser225Stop, Gly239Stop, Lys393Stop, Leu203Stop, Gly44Arg, Glu257Stop | Point mutations in human CCRF-CEM leukemia cells. | Human CCRF-CEM leukemia cells resistant to methotrexate and other anti-folates (e.g. GW1843, PT523, pemetrexed, trimetrexate, edatrexate)
|
Not determined |
Source:
Note:
These leukemia samples were collected during the time of diagnosis and approximately 10% of the samples were at the time of disease recurrence.
These variants are likely from somatic cells although the possibility that the variants are from normal cell DNA (germline DNA) that contaminated the tumor sample cannot be ruled out.
Important variants
(For full mapping information, see http://www.pharmgkb.org/search/annotatedGene/slc19a1/variant.jsp) The SLC19A1 gene is polymorphic in humans. The effect of the variants in SLC19A1 has been widely studied in various clinical conditions where folate transport, synthesis and metabolism pathways are involved.
SLC19A1: Arg27His; 80G>A (rs1051266)
SLC19A1: 5’-UTR variant, −43T>C, (rs1131596)
SLC19A1: Pro232Pro; 6318C>T (rs12659)
SLC19A1: 3’-UTR; 2606G>T (rs1051296)
SLC19A1: 3'UTR, 2522C>T (rs1051298)
1) SLC19A1: Arg27His; 80G>A (rs1051266)
The most extensively studied variant of SLC19A1 gene is 80G>A (rs1051266), a common non-synonymous polymorphism in exon 2 that results in substitution of a histidine for an arginine at residue 27 in the protein sequence. The estimated genotype frequencies among Caucasians were found to be: GG=0.29, GA=0.473, AA=0.237 [30]. The frequency of allele A was 0.473 in Caucasians, 0.564 in African Americans, and 0.472 in Hispanics. This variant has been widely studied for its function in transport uptake as well as its association with risk to diseases and drug response and toxicity. Below we summarize examples of these associations with this important SLC19A1 variant, Arg27His.
Association with plasma folate levels
This common nonsynonymous variant of SLC19A1 80G>A was associated with plasma folate levels in which individuals who were homozygous AA had higher plasma folate levels in comparison to individuals who carried the G allele [31]. This association was not observed in other studies [32, 33]. One study observed an association of SLC19A1 80G>A and folate levels in red blood cells, and this association is greater in women than in men [33].
Association with risk of common birth defects
Since folic acid and vitamin B12 are very important in reducing the occurrences of common birth defects such as neural tube defects, or NTD, the genetic variants in SLC19A1 and other folate pathway genes have been investigated for their roles in common birth defects. One study showed that in Italians, the G allele of variant G80A was observed at higher frequencies in affected children with neural tube defects (NTD), their mothers, and their fathers [34]. Another study investigated the interaction between use of periconceptional folate supplementation in mothers and the risk of spina bifida in their infants as a function of the genotype at nucleotide position 80 of SLC19A1 in the infants [35]. It was found that infants born from mothers who did not take folate during periconception were at higher risk of having spina bifida if they carried the GG genotype (OR=2.4) compared to infants with the AA genotype. On the other hand, infants whose mothers took folate during periconception had a decreased risk of spina bifida if they carried the GG genotype compared to infants with the AA genotype. Although the findings did not reach statistical significance, this study reveals a potentially interesting gene-nutrient interaction involving folate use in mothers and SLC19A1 genotype at nucleotide position 80 in infants and risk of spina bifida. The G80A variant has also been studied in congenital defects, such as congenital heart disease (CHD) and cleft palate. One study showed that infants born from mothers who did not use folate supplementation were at significantly higher risk of CHD (OR=2.94) compared to infants whose mothers took folate during periconception. In addition, the CHD risk was significantly higher in infants with the GG (OR=4.03) and GA (OR=4.14) genotypes compared to infants with the AA genotype, if their mothers did not use folate supplementation. No significant association was found between SLC19A1 G80 genotype or maternal folic acid supplementation and the risks of cleft palate [36].
Association with other risks affected by folate levels
There are several biosynthetic pathways in humans that require folate. As a result, any changes to the folate bioavailability could influence human health. A study involving 156 patients showed that the A allele had a significant protective effect against thrombosis (OR = 0.56) [37].
Association with methotrexate transport
The functional effect of the G80A polymorphism was assessed in transport-impaired K562 cells transfected with the reference Arg27 or variant His27-RFC1 proteins [38]. The variant protein, Arg27His, transported methotrexate and 5-formyl tetrahydrofolate similarly compared to reference RFC1. Although the kinetics of methotrexate transport by Arg27-RFC1 protein were not significantly different compared to K562 cells transfected with His27-RFC1, minor differences (~ 2-fold) were detected between the Arg27-RFC1 and His27-RFC1 proteins in the interaction affinity (Ki) of other anti-folates, including Tomudex, 5,10-dideazatetrahydrofolate, GW1843U89, 10-ethyl-10-deazaaminopterin, and 5-formyl tetrahydrofolate. The Ki for these other anti-folates in His27-RFC1 were ~ 2-fold lower although there were no significant differences between the K562 cells transfected with Arg27-RFC1 and His27-RFC1 with their sensitivities to growth inhibition by methotrexate and other anti-folates. The results suggested that there were at most minor functional differences between Arg27- and His27-RFC1 in terms of substrate affinities and/or transport efficiencies.
Association with methotrexate plasma levels and response
Altered cellular uptake of methotrexate may have an effect on methotrexate plasma level and thus hamper the efficacy and toxicities of methotrexate. A clinical study assessed whether there was an association of this variant Arg27His with methotrexate plasma level and clinical outcome in childhood acute lymophoblastic leukemias [39]. The result showed that children with the AA genotype had more adverse events and worse overall prognosis than patients with the GG genotype. In addition, it was determined that patients carrying the AA genotype had higher methotrexate plasma levels (P =0.004) than patients with either the GG or GA genotypes. Another clinical study was conduced in patients treated with weekly low-dose methotrexate for rheumatoid arthritis [40]. The result showed that the AA genotype was associated with 3.4-fold higher level of methotrexate polyglutamates (the active metabolites of methotrexate) compared to those with the 80GG and 80GA (OR 95% CI 1.4 – 8.4; p=0.007). This result is supported by another study, where they showed that patients with AA genotype have a 3.32-fold higher probability of remission of rheutmatoid arthritis symptoms [41]. Genetic variants in the candidate genes of the folate pathway have also been examined for their role in drug response. One of the studies describes the association of polymorphisms in folate pathway with the response of methotrexate and sulfasalazine combination regimens in early rheumatoid arthritis patients [42]. In this study (n=98 Caucasians), a gene-gene interaction between SLC19A1 80A allele and MTR (5-methyltetrahydrofolate-homocysteine methyltransferase) 2756A (rs1805087) is observed with response to the therapy. The result showed that the allele combinations associated with responders are MTR2756A allele in combination with SLC19A1 80A allele (multivariate analysis, p=0.0002). Recently, Gregers et al. reported the influence of this variant G80A on the risk of methotrexate relapse and toxicity [43]. This study consisting of 500 children with ALL found that the AA variant was associated with a 50% better chance of staying in remission compared with GG or GA variants.
Association with methotrexate toxicities
Given the importance of SLC19A1 G80A in methotrexate plasma level and response as discussed above, additional studies have been carried out to assess the effect of this variant with methotrexate toxicities in ALL patients who are treated with high-dose methotrexate. A large retrospective candidate gene study involving 240 ALL patients, this variant G80A is associated with methotrexate gastrointestinal toxicity [44]. In addition, a recent study by Gregers et al. reported that children with ALL (a total of 182 children with toxicity phenotype information) who have AA variant has higher degree of bone marrow toxicity and a higher degree of liver toxicity [43].
Association with risk of cancer
Genes involved with folate uptake and distribution have been investigated for different types of cancer risk including breast, prostate, colorectal and lymphoma [45–48]. Most of them have found no significant association of this variant with the cancer risk. However, a recent case-control study involving Chinese population revealed significant association of 80AA with increased risk of esophageal cancer (n=216) and gastric cancer (n=633) with adjusted odds ratio (OR) of 1.80 (95% confidence interval = 1.29–2.51) and 1.59 (95% confidence interval = 1.25–2.02) respectively [49]. In addition, a case-control study involving 245 pediatric acute lymphobastoid leukemia patients and 500 controls has found a gene-gene interaction between SLC19A1 and nicotinamide N-methyltransferase (NNMT) [50], where subjects with SLC19A1 80AA/NNMT IVS – 151CT + TT genotype had a 4.2 fold increase in ALL risk (p=0.001).
2) SLC19A1: 5’-UTR variant, −43T>C, (rs1131596)
This common variant in the 5’-UTR region of SLC19A1 is found in all HapMap populations. A clinical study involving 106 rheumatoid arthritis patients who received methotrexate treatment has found this variant is associated with lower RFC1 protein expression [51] and it is predicted that this regulatory region shows putative consensus sequence of the AP1 transcription factor. This variant is also associated with red blood cell folate levels, with the C allele associated with low red blood cell folate levels. However, this regulatory variant is not sufficient to predict patient response to MTX therapy or toxicity [51].
3) SLC19A1: Pro232Pro; 6318C>T (rs12659)
In a study which involved 122 lung cancer cases and 122 matched controls in Xuan Wei, China, where the incidence of lung cancer mortality is high due to indoor smoky coal emissions [52], the group found that the synonymous variant in SLC19A1 Pro232Pro (rs12659) is associated with an increased risk of lung cancer in this population (OR=1.83, 95% CI = 1.02 – 3.28). The effect of this variant Pro232Pro, as well as other polymorphisms in genes involved in one-carbon metabolic pathway which controls nucleotide synthesis and DNA methylation [53], was assessed for their effect on non-Hodgkin lymphoma (NHL) among over 1000 cases and 949 population-based controls. However, the variant Pro232Pro did not show any protective association. A positive association of this variant Pro232Pro is implicated in a study involving cervical carcinoma patients where platinum-based chemotherapy was used for their treatment [54]. In this study, the probability of response was higher in patients with the variant allele T (P=0.032).
4) SLC19A1: 3’-UTR ; 2606G>T (rs1051296)
This variant in the 3’-UTR region of SLC19A1 showed high allele frequencies (40 – 50%) in all HapMap populations. Potentially, a SNP located within the 3’-untranslated region (UTR), which contains regulatory sequences and binding sites for other molecules that could alter the stability of the mRNA transcript of the gene. However, this SNP has not been functionally characterized although this SNP has been genotyped and assessed for its effect on non-Hodgkin lymphoma (NHL) among over 1000 cases and 949 population-based controls [53]. There is no significant association of this variant with risk of NHL.
5) SLC19A1: 3'UTR, 2522C>T (rs1051298)
Similarly to rs1051296, this variant in the 3’-UTR region of SLC19A1 (rs1051298) showed high allele frequencies (40 – 50%) in all HapMap populations. An analog of methotrexate called pemetrexed, which is transported into the cells by SLC19A1, has recently been approved for its indication in non-small cell lung cancer. Forty-eight patients enrolled in the Phase II clinical trial (NCCTG and SWOG study N0426) for pemetrexed plus bevacizumab in advanced non-small cell lung cancer, were genotyped for the this SNP in SLC19A1 as well as other drug metabolizing enzymes [55]. The result from this study showed that the variant T allele is correlated with 3-month progression-free status (P=0.01) and with median progression-free survival (CC v CT v TT: 5.7 v 2.8 v 4.7 months, respectively; log-rank test, P=0.05).
Conclusion
SLC19A1 is ubiquitously expressed in human tissues and is recognized for its role in the transport of folates and anti-folates drugs. Its role together with other genes that are involved in folate synthesis and metabolism have important implications in governing folate homeostasis and thus in preventing diseases such as neural tube defects and other birth defects. Variants in the SLC19A1 gene have been found to be correlated with various cancers and with variable response to methotrexate and related compounds for rheumatoid arthritis and cancer. The common non-synonymous variant Arg27His has been widely studied and some studies have showed significant correlation of the variants with disease risk and/or cancer. The significant associations have failed to be replicated in some studies, suggesting that the overall contributions to the phenotype may not be largely affected and cannot be explained by this variant alone. Besides disease risk, the effect of this common variant has been investigated in pharmacogenomics studies related to methotrexate response and toxicity. The overall evidence suggests that the A allele of this variant Arg27His (G80>A) is associated with protective effect in MTX response yet higher toxicity to high dose MTX treatment [42–44]. However, this variant did not reach genomewide significant in the recent genomewide association study on methotrexate clearance and toxicity [56], suggesting that genes other than SLC19A1 gene may be the major contributing factor for methotrexate response and toxicity. Large prospective study will be required to determine the effect of genetic variant in SLC19A1 on risk of methotrexate treatment failure as well as determining whether dosing adjustment to methotrexate according to SLC19A1 genotype can improve overall treatment efficacy are important questions to be addressed. Although some rare non-synonymous variants (≤1% MAF) have been reported through sequencing effort in healthy subjects, resistant cell lines and tumor samples, the effects of these variants in clinical outcome are yet to be determined.
Acknowledgements
PharmGKB is supported by the NIH/NIGMS UO1GM61374. PMT (Pharmacogenetics of Membrane Transporters) is supported by NIH/NIGMS Pharmacogenetics Research Network (PGRN; U01GM61390).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Additional information, including detailed mapping information for SLC19A1 gene (PA327), variants and lists of linked drugs and diseases is presented at http://www.pharmgkb.org/search/annotatedGene/slc19a1/index.jsp.
References
- 1.Smulders YM, Stehouwer CD. Folate metabolism and cardiovascular disease. Semin Vasc Med. 2005;5:87–97. doi: 10.1055/s-2005-872395. [DOI] [PubMed] [Google Scholar]
- 2.Prasad PD, Ramamoorthy S, Leibach FH, Ganapathy V. Molecular cloning of the human placental folate transporter. Biochem Biophys Res Commun. 1995;206:681–687. doi: 10.1006/bbrc.1995.1096. [DOI] [PubMed] [Google Scholar]
- 3.Nguyen TT, Dyer DL, Dunning DD, Rubin SA, Grant KE, Said HM. Human intestinal folate transport: cloning, expression, and distribution of complementary RNA. Gastroenterology. 1997;112:783–791. doi: 10.1053/gast.1997.v112.pm9041240. [DOI] [PubMed] [Google Scholar]
- 4.Wong SC, Proefke SA, Bhushan A, Matherly LH. Isolation of human cDNAs that restore methotrexate sensitivity and reduced folate carrier activity in methotrexate transport-defective Chinese hamster ovary cells. J Biol Chem. 1995;270:17468–17475. doi: 10.1074/jbc.270.29.17468. [DOI] [PubMed] [Google Scholar]
- 5.Williams FM, Flintoff WF. Isolation of a human cDNA that complements a mutant hamster cell defective in methotrexate uptake. J Biol Chem. 1995;270:2987–2992. doi: 10.1074/jbc.270.7.2987. [DOI] [PubMed] [Google Scholar]
- 6.Dixon KH, Lanpher BC, Chiu J, Kelley K, Cowan KH. A novel cDNA restores reduced folate carrier activity and methotrexate sensitivity to transport deficient cells. J Biol Chem. 1994;269:17–20. [PubMed] [Google Scholar]
- 7.Chiao JH, Roy K, Tolner B, Yang CH, Sirotnak FM. RFC-1 gene expression regulates folate absorption in mouse small intestine. J Biol Chem. 1997;272:11165–11170. doi: 10.1074/jbc.272.17.11165. [DOI] [PubMed] [Google Scholar]
- 8.Zhao R, Gao F, Goldman ID. Reduced folate carrier transports thiamine monophosphate: an alternative route for thiamine delivery into mammalian cells. Am J Physiol Cell Physiol. 2002;282:C1512–C1517. doi: 10.1152/ajpcell.00547.2001. [DOI] [PubMed] [Google Scholar]
- 9.Matherly LH, Hou Z, Deng Y. Human reduced folate carrier: translation of basic biology to cancer etiology and therapy. Cancer Metastasis Rev. 2007;26:111–128. doi: 10.1007/s10555-007-9046-2. [DOI] [PubMed] [Google Scholar]
- 10.Ifergan I, Jansen G, Assaraf YG. The reduced folate carrier (RFC) is cytotoxic to cells under conditions of severe folate deprivation. RFC as a double edged sword in folate homeostasis. J Biol Chem. 2008;283:20687–20695. doi: 10.1074/jbc.M802812200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Williams FM, Flintoff WF. Structural organization of the human reduced folate carrier gene: evidence for 5' heterogeneity in lymphoblast mRNA. Somat Cell Mol Genet. 1998;24:143–156. doi: 10.1023/b:scam.0000007117.50428.63. [DOI] [PubMed] [Google Scholar]
- 12.Tolner B, Roy K, Sirotnak FM. Structural analysis of the human RFC-1 gene encoding a folate transporter reveals multiple promoters and alternatively spliced transcripts with 5' end heterogeneity. Gene. 1998;211:331–341. doi: 10.1016/s0378-1119(98)00123-1. [DOI] [PubMed] [Google Scholar]
- 13.Whetstine JR, Witt TL, Matherly LH. The human reduced folate carrier gene is regulated by the AP2 and sp1 transcription factor families and a functional 61-base pair polymorphism. J Biol Chem. 2002;277:43873–43880. doi: 10.1074/jbc.M208296200. [DOI] [PubMed] [Google Scholar]
- 14.Whetstine JR, Flatley RM, Matherly LH. The human reduced folate carrier gene is ubiquitously and differentially expressed in normal human tissues: identification of seven non-coding exons and characterization of a novel promoter. Biochem J. 2002;367:629–640. doi: 10.1042/BJ20020512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Patterson D, Graham C, Cherian C, Matherly LH. A humanized mouse model for the reduced folate carrier. Mol Genet Metab. 2008;93:95–103. doi: 10.1016/j.ymgme.2007.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Tse A, Brigle K, Taylor SM, Moran RG. Mutations in the reduced folate carrier gene which confer dominant resistance to 5,10-dideazatetrahydrofolate. J Biol Chem. 1998;273:25953–25960. doi: 10.1074/jbc.273.40.25953. [DOI] [PubMed] [Google Scholar]
- 17.Gorlick R, Goker E, Trippett T, Steinherz P, Elisseyeff Y, Mazumdar M, Flintoff WF, Bertino JR. Defective transport is a common mechanism of acquired methotrexate resistance in acute lymphocytic leukemia and is associated with decreased reduced folate carrier expression. Blood. 1997;89:1013–1018. [PubMed] [Google Scholar]
- 18.Wong SC, Zhang L, Proefke SA, Hukku B, Matherly LH. Gene amplification and increased expression of the reduced folate carrier in transport elevated K562 cells. Biochem Pharmacol. 1998;55:1135–1138. doi: 10.1016/s0006-2952(97)00639-4. [DOI] [PubMed] [Google Scholar]
- 19.Wong SC, Zhang L, Witt TL, Proefke SA, Bhushan A, Matherly LH. Impaired membrane transport in methotrexate-resistant CCRF-CEM cells involves early translation termination and increased turnover of a mutant reduced folate carrier. J Biol Chem. 1999;274:10388–10394. doi: 10.1074/jbc.274.15.10388. [DOI] [PubMed] [Google Scholar]
- 20.Yang R, Li WW, Hoang BH, Kim H, Banerjee D, Kheradpour A, Healey JH, Meyers PA, Bertino JR, Gorlick R. Quantitative correlation between promoter methylation and messenger RNA levels of the reduced folate carrier. BMC Cancer. 2008;8:124. doi: 10.1186/1471-2407-8-124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Charasson V, Hillaire-Buys D, Solassol I, Laurand-Quancard A, Pinguet F, Le Morvan V, Robert J. Involvement of gene polymorphisms of the folate pathway enzymes in gene expression and anticancer drug sensitivity using the NCI-60 panel as a model. Eur J Cancer. 2009;45:2391–2401. doi: 10.1016/j.ejca.2009.05.013. [DOI] [PubMed] [Google Scholar]
- 22.Belkov VM, Krynetski EY, Schuetz JD, Yanishevski Y, Masson E, Mathew S, Raimondi S, Pui CH, Relling MV, Evans WE. Reduced folate carrier expression in acute lymphoblastic leukemia: a mechanism for ploidy but not lineage differences in methotrexate accumulation. Blood. 1999;93:1643–1650. [PubMed] [Google Scholar]
- 23.Kager L, Cheok M, Yang W, Zaza G, Cheng Q, Panetta JC, Pui CH, Downing JR, Relling MV, Evans WE. Folate pathway gene expression differs in subtypes of acute lymphoblastic leukemia and influences methotrexate pharmacodynamics. J Clin Invest. 2005;115:110–117. doi: 10.1172/JCI22477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Gifford AJ, Haber M, Witt TL, Whetstine JR, Taub JW, Matherly LH, Norris MD. Role of the E45K-reduced folate carrier gene mutation in methotrexate resistance in human leukemia cells. Leukemia. 2002;16:2379–2387. doi: 10.1038/sj.leu.2402655. [DOI] [PubMed] [Google Scholar]
- 25.Jansen G, Mauritz R, Drori S, Sprecher H, Kathmann I, Bunni M, Priest DG, Noordhuis P, Schornagel JH, Pinedo HM, et al. A structurally altered human reduced folate carrier with increased folic acid transport mediates a novel mechanism of antifolate resistance. J Biol Chem. 1998;273:30189–30198. doi: 10.1074/jbc.273.46.30189. [DOI] [PubMed] [Google Scholar]
- 26.Drori S, Jansen G, Mauritz R, Peters GJ, Assaraf YG. Clustering of mutations in the first transmembrane domain of the human reduced folate carrier in GW1843U89-resistant leukemia cells with impaired antifolate transport and augmented folate uptake. J Biol Chem. 2000;275:30855–30863. doi: 10.1074/jbc.M003988200. [DOI] [PubMed] [Google Scholar]
- 27.Kaufman Y, Ifergan I, Rothem L, Jansen G, Assaraf YG. Coexistence of multiple mechanisms of PT523 resistance in human leukemia cells harboring 3 reduced folate carrier alleles: transcriptional silencing, inactivating mutations, and allele loss. Blood. 2006;107:3288–3294. doi: 10.1182/blood-2005-10-4048. [DOI] [PubMed] [Google Scholar]
- 28.Kaufman Y, Drori S, Cole PD, Kamen BA, Sirota J, Ifergan I, Arush MW, Elhasid R, Sahar D, Kaspers GJ, et al. Reduced folate carrier mutations are not the mechanism underlying methotrexate resistance in childhood acute lymphoblastic leukemia. Cancer. 2004;100:773–782. doi: 10.1002/cncr.20018. [DOI] [PubMed] [Google Scholar]
- 29.Yang R, Sowers R, Mazza B, Healey JH, Huvos A, Grier H, Bernstein M, Beardsley GP, Krailo MD, Devidas M, et al. Sequence alterations in the reduced folate carrier are observed in osteosarcoma tumor samples. Clin Cancer Res. 2003;9:837–844. [PubMed] [Google Scholar]
- 30.Rady PL, Szucs S, Matalon RK, Grady J, Hudnall SD, Kellner LH, Nitowsky H. Genetic polymorphism (G80A) of reduced folate carrier gene in ethnic populations. Mol Genet Metab. 2001;73:285–286. doi: 10.1006/mgme.2001.3197. [DOI] [PubMed] [Google Scholar]
- 31.Chango A, Emery-Fillon N, de Courcy GP, Lambert D, Pfister M, Rosenblatt DS, Nicolas JP. A polymorphism (80G->A) in the reduced folate carrier gene and its associations with folate status and homocysteinemia. Mol Genet Metab. 2000;70:310–315. doi: 10.1006/mgme.2000.3034. [DOI] [PubMed] [Google Scholar]
- 32.Devlin AM, Clarke R, Birks J, Evans JG, Halsted CH. Interactions among polymorphisms in folate-metabolizing genes and serum total homocysteine concentrations in a healthy elderly population. Am J Clin Nutr. 2006;83:708–713. doi: 10.1093/ajcn.83.3.708. [DOI] [PubMed] [Google Scholar]
- 33.Stanislawska-Sachadyn A, Mitchell LE, Woodside JV, Buckley PT, Kealey C, Young IS, Scott JM, Murray L, Boreham CA, McNulty H, et al. The reduced folate carrier (SLC19A1) c.80G>A polymorphism is associated with red cell folate concentrations among women. Ann Hum Genet. 2009;73:484–491. doi: 10.1111/j.1469-1809.2009.00529.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.De Marco P, Calevo MG, Moroni A, Arata L, Merello E, Cama A, Finnell RH, Andreussi L, Capra V. Polymorphisms in genes involved in folate metabolism as risk factors for NTDs. Eur J Pediatr Surg. 2001;11 Suppl 1:S14–S17. doi: 10.1055/s-2001-19739. [DOI] [PubMed] [Google Scholar]
- 35.Shaw GM, Lammer EJ, Zhu H, Baker MW, Neri E, Finnell RH. Maternal periconceptional vitamin use, genetic variation of infant reduced folate carrier (A80G), and risk of spina bifida. Am J Med Genet. 2002;108:1–6. doi: 10.1002/ajmg.10195. [DOI] [PubMed] [Google Scholar]
- 36.Pei L, Zhu H, Zhu J, Ren A, Finnell RH, Li Z. Genetic variation of infant reduced folate carrier (A80G) and risk of orofacial defects and congenital heart defects in China. Ann Epidemiol. 2006;16:352–356. doi: 10.1016/j.annepidem.2005.02.014. [DOI] [PubMed] [Google Scholar]
- 37.Yates Z, Lucock M. G80A reduced folate carrier SNP modulates cellular uptake of folate and affords protection against thrombosis via a non homocysteine related mechanism. Life Sci. 2005;77:2735–2742. doi: 10.1016/j.lfs.2005.02.029. [DOI] [PubMed] [Google Scholar]
- 38.Whetstine JR, Gifford AJ, Witt T, Liu XY, Flatley RM, Norris M, Haber M, Taub JW, Ravindranath Y, Matherly LH. Single nucleotide polymorphisms in the human reduced folate carrier: characterization of a high-frequency G/A variant at position 80 and transport properties of the His(27) and Arg(27) carriers. Clin Cancer Res. 2001;7:3416–3422. [PubMed] [Google Scholar]
- 39.Laverdiere C, Chiasson S, Costea I, Moghrabi A, Krajinovic M. Polymorphism G80A in the reduced folate carrier gene and its relationship to methotrexate plasma levels and outcome of childhood acute lymphoblastic leukemia. Blood. 2002;100:3832–3834. doi: 10.1182/blood.V100.10.3832. [DOI] [PubMed] [Google Scholar]
- 40.Dervieux T, Kremer J, Lein DO, Capps R, Barham R, Meyer G, Smith K, Caldwell J, Furst DE. Contribution of common polymorphisms in reduced folate carrier and gamma-glutamylhydrolase to methotrexate polyglutamate levels in patients with rheumatoid arthritis. Pharmacogenetics. 2004;14:733–739. doi: 10.1097/00008571-200411000-00004. [DOI] [PubMed] [Google Scholar]
- 41.Drozdzik M, Rudas T, Pawlik A, Gornik W, Kurzawski M, Herczynska M. Reduced folate carrier-1 80G>A polymorphism affects methotrexate treatment outcome in rheumatoid arthritis. Pharmacogenomics J. 2007;7:404–407. doi: 10.1038/sj.tpj.6500438. [DOI] [PubMed] [Google Scholar]
- 42.James HM, Gillis D, Hissaria P, Lester S, Somogyi AA, Cleland LG, Proudman SM. Common polymorphisms in the folate pathway predict efficacy of combination regimens containing methotrexate and sulfasalazine in early rheumatoid arthritis. J Rheumatol. 2008;35:562–571. [PubMed] [Google Scholar]
- 43.Gregers J, Christensen IJ, Dalhoff K, Lausen B, Schroeder H, Rosthoej S, Carlsen N, Schmiegelow K, Peterson C. The association of reduced folate carrier 80G>A polymorphism to outcome in childhood acute lymphoblastic leukemia interacts with chromosome 21 copy number. Blood. 2010;115:4671–4677. doi: 10.1182/blood-2010-01-256958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kishi S, Cheng C, French D, Pei D, Das S, Cook EH, Hijiya N, Rizzari C, Rosner GL, Frudakis T, et al. Ancestry and pharmacogenetics of antileukemic drug toxicity. Blood. 2007;109:4151–4157. doi: 10.1182/blood-2006-10-054528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Figueiredo JC, Levine AJ, Lee WH, Conti DV, Poynter JN, Campbell PT, Duggan D, Lewinger JP, Martinez ME, Ulrich CM, et al. Genes involved with folate uptake and distribution and their association with colorectal cancer risk. Cancer Causes Control. 2009 doi: 10.1007/s10552-009-9489-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Collin SM, Metcalfe C, Zuccolo L, Lewis SJ, Chen L, Cox A, Davis M, Lane JA, Donovan J, Smith GD, et al. Association of folate-pathway gene polymorphisms with the risk of prostate cancer: a population-based nested case-control study, systematic review, and meta-analysis. Cancer Epidemiol Biomarkers Prev. 2009;18:2528–2539. doi: 10.1158/1055-9965.EPI-09-0223. [DOI] [PubMed] [Google Scholar]
- 47.Lee KM, Lan Q, Kricker A, Purdue MP, Grulich AE, Vajdic CM, Turner J, Whitby D, Kang D, Chanock S, et al. One-carbon metabolism gene polymorphisms and risk of non-Hodgkin lymphoma in Australia. Hum Genet. 2007;122:525–533. doi: 10.1007/s00439-007-0431-2. [DOI] [PubMed] [Google Scholar]
- 48.Lissowska J, Gaudet MM, Brinton LA, Chanock SJ, Peplonska B, Welch R, Zatonski W, Szeszenia-Dabrowska N, Park S, Sherman M, Garcia-Closas M. Genetic polymorphisms in the one-carbon metabolism pathway and breast cancer risk: a population-based case-control study and meta-analyses. Int J Cancer. 2007;120:2696–2703. doi: 10.1002/ijc.22604. [DOI] [PubMed] [Google Scholar]
- 49.Wang L, Chen W, Wang J, Tan Y, Zhou Y, Ding W, Hua Z, Shen J, Xu Y, Shen H. Reduced folate carrier gene G80A polymorphism is associated with an increased risk of gastroesophageal cancers in a Chinese population. Eur J Cancer. 2006;42:3206–3211. doi: 10.1016/j.ejca.2006.04.022. [DOI] [PubMed] [Google Scholar]
- 50.de Jonge R, Tissing WJ, Hooijberg JH, Jansen G, Kaspers GJ, Lindemans J, Peters GJ, Pieters R. Polymorphisms in folate-related genes and risk of pediatric acute lymphoblastic leukemia. Blood. 2009;113:2284–2289. doi: 10.1182/blood-2008-07-165928. [DOI] [PubMed] [Google Scholar]
- 51.Chatzikyriakidou A, Georgiou I, Voulgari PV, Papadopoulos CG, Tzavaras T, Drosos AA. Transcription regulatory polymorphism −43T>C in the 5'-flanking region of SLC19A1 gene could affect rheumatoid arthritis patient response to methotrexate therapy. Rheumatol Int. 2007;27:1057–1061. doi: 10.1007/s00296-007-0339-0. [DOI] [PubMed] [Google Scholar]
- 52.Shen M, Rothman N, Berndt SI, He X, Yeager M, Welch R, Chanock S, Caporaso N, Lan Q. Polymorphisms in folate metabolic genes and lung cancer risk in Xuan Wei, China. Lung Cancer. 2005;49:299–309. doi: 10.1016/j.lungcan.2005.04.002. [DOI] [PubMed] [Google Scholar]
- 53.Lim U, Wang SS, Hartge P, Cozen W, Kelemen LE, Chanock S, Davis S, Blair A, Schenk M, Rothman N, Lan Q. Gene-nutrient interactions among determinants of folate and one-carbon metabolism on the risk of non-Hodgkin lymphoma: NCI-SEER case-control study. Blood. 2007;109:3050–3059. doi: 10.1182/blood-2006-07-034330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Chung HH, Kim MK, Kim JW, Park NH, Song YS, Kang SB, Lee HP. XRCC1 R399Q polymorphism is associated with response to platinum-based neoadjuvant chemotherapy in bulky cervical cancer. Gynecol Oncol. 2006;103:1031–1037. doi: 10.1016/j.ygyno.2006.06.016. [DOI] [PubMed] [Google Scholar]
- 55.Adjei AA, Mandrekar SJ, Dy GK, Molina JR, Adjei AA, Gandara DR, Ziegler KL, Stella PJ, Rowland KM, Jr, Schild SE, Zinner RG. Phase II trial of pemetrexed plus bevacizumab for second-line therapy of patients with advanced non-small-cell lung cancer: NCCTG and SWOG study N0426. J Clin Oncol. 2010;28:614–619. doi: 10.1200/JCO.2009.23.6406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Trevino LR, Shimasaki N, Yang W, Panetta JC, Cheng C, Pei D, Chan D, Sparreboom A, Giacomini KM, Pui CH et al. Germline genetic variation in an organic anion transporter polypeptide associated with methotrexate pharmacokinetics and clinical effects. J Clin Oncol. 2009;27:5972–5978. doi: 10.1200/JCO.2008.20.4156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Shimasaki N, Mori T, Samejima H, Sato R, Shimada H, Yahagi N, Torii C, Yoshihara H, Tanigawara Y, Takahashi T, Kosaki K. Effects of methylenetetrahydrofolate reductase and reduced folate carrier 1 polymorphisms on high-dose methotrexate-induced toxicities in children with acute lymphoblastic leukemia or lymphoma. J Pediatr Hematol Oncol. 2006;28:64–68. doi: 10.1097/01.mph.0000198269.61948.90. [DOI] [PubMed] [Google Scholar]
- 58.Zhao R, Assaraf YG, Goldman ID. A mutated murine reduced folate carrier (RFC1) with increased affinity for folic acid, decreased affinity for methotrexate, and an obligatory anion requirement for transport function. J Biol Chem. 1998;273:19065–19071. doi: 10.1074/jbc.273.30.19065. [DOI] [PubMed] [Google Scholar]
- 59.Rothem L, Ifergan I, Kaufman Y, Priest DG, Jansen G, Assaraf YG. Resistance to multiple novel antifolates is mediated via defective drug transport resulting from clustered mutations in the reduced folate carrier gene in human leukaemia cell lines. Biochem J. 2002;367:741–750. doi: 10.1042/BJ20020801. [DOI] [PMC free article] [PubMed] [Google Scholar]