Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Leukemia ; 16(12): 2379-87, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12454742

ABSTRACT

Resistance to the antifolate methotrexate (MTX) can cause treatment failure in childhood acute lymphoblastic leukemia (ALL). This may result from defective MTX accumulation due to alterations in the human reduced folate carrier (hRFC) gene. We have identified an hRFC gene point mutation in a transport-defective CCRF-CEM human T-ALL cell line resulting in a lysine to glutamic acid substitution at codon 45 (E45K), which has been identified in other antifolate-resistant sublines (JBC 273:30 189, 1998; JBC 275:30 855, 2000). To characterize the role of this mutation in MTX resistance, transfection experiments were performed using hRFC-null CCRF-CEM cells. E45K transfectants demonstrated an initial rate of MTX influx that was approximately 0.5-fold that of CCRF-CEM cells, despite marked protein overexpression. Cytotoxicity studies revealed partial reversal of MTX and raltitrexed resistance in E45K transfectants, while trimetrexate resistance was significantly increased. Kinetic analysis indicated only minor differences in MTX kinetics between wild-type and E45K hRFCs, however, K(i)s for folic acid and 5-formyltetrahydrofolate were markedly reduced for E45K hRFC. This was paralleled by increased folic acid transport and reduced synthesis of MTX polyglutamates. Collectively, the results demonstrate that expression of E45K hRFC leads to increased MTX resistance due to decreased membrane transport and, secondarily, from alterations in binding affinities and transport of folate substrates. However, despite these findings, we could find no evidence of this mutation in 121 childhood ALL samples, suggesting that it does not contribute to clinical MTX resistance in this disease.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Carrier Proteins/genetics , Drug Resistance, Neoplasm/genetics , Leukemia/drug therapy , Membrane Transport Proteins , Methotrexate/pharmacokinetics , Point Mutation , Amino Acid Substitution , Bone Marrow Cells/pathology , Carrier Proteins/physiology , Child , Folic Acid/pharmacokinetics , Humans , Kinetics , Leukemia/genetics , Leukemia/pathology , Protein Structure, Tertiary , Reduced Folate Carrier Protein , Transfection , Tumor Cells, Cultured
2.
Clin Cancer Res ; 7(11): 3416-22, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11705857

ABSTRACT

The presence of sequence variants in the human reduced folate carrier (hRFC) was assessed in leukemia blasts from children with acute lymphoblastic leukemia (ALL) and in normal peripheral blood specimens. A CATG frame shift insertion at position 191 was detected in 10-60% of hRFC transcripts from 10 of 16 ALL specimens, by RFLP analysis and direct sequencing of hRFC cDNAs. In genomic DNAs prepared from 105 leukemia (n = 54) and non-leukemia (n = 51) specimens, PCR amplifications and direct sequencing of exon 3 identified a high-frequency G to A single nucleotide polymorphism at position 80 that resulted in a change of arginine-27 to histidine-27. The allelic frequencies of G/A80 were nearly identical for the non-leukemia (42.2% CGC and 57.8% CAC) and leukemia (40.7% CGC and 59.3% CAC) genomic DNAs. In cDNAs prepared from 10 of these ALL patients, identical allelic frequencies (40 and 60%, respectively) were recorded. In up to 62 genomic DNAs, hRFC-coding exons 4-7 were PCR-amplified and sequenced. A high-abundance C/T696 polymorphism was detected with nearly identical frequencies for both alleles, and a heterozygous C/A1242 sequence variant was identified in two ALL specimens. Both C/T696 and C/A1242 were phenotypically silent. In transport assays with [(3)H]methotrexate and [(3)H]5-formyl tetrahydrofolate, nearly identical uptake rates were measured for the arginine-27- and histidine-27-hRFC proteins expressed in transport-impaired K562 cells. Although there were no significant differences between the kinetic parameters for methotrexate transport for the hRFC forms, minor (approximately 2-fold) differences were measured in the K(i)s for other substrates including Tomudex, 5,10-dideazatetrahydrofolate, GW1843U89, and 10-ethyl-10-deazaaminopterin and for 5-formyl tetrahydrofolate.


Subject(s)
Carrier Proteins/genetics , Membrane Transport Proteins , Polymorphism, Single Nucleotide , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Amino Acid Substitution , B-Lymphocytes/metabolism , Base Sequence , Biological Transport/genetics , Child , DNA Mutational Analysis , DNA, Complementary/chemistry , DNA, Complementary/genetics , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Gene Frequency , Humans , K562 Cells , Methotrexate/pharmacokinetics , Mutagenesis, Insertional , Plasmids/genetics , Point Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Reduced Folate Carrier Protein , Stem Cells/metabolism , Transfection
3.
J Biol Chem ; 276(12): 8713-9, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11106643

ABSTRACT

The relationship between loss of functional p53 and human reduced folate carrier (hRFC) levels and function was examined in REH lymphoblastic leukemia cells, which express wild type p53, and in p53-null K562 cells (K562(pTet-on/p53)) engineered to express wild type p53 under control of a tetracycline-inducible promoter. Activation of p53 in REH cells by treatment with daunorubicin was accompanied by decreased ( approximately 5-fold) levels of hRFC transcripts and methotrexate transport. Treatment of K562(pTet-on/p53) cells with doxycycline resulted in a dose-dependent expression of p53 protein and transcripts, increased p21 protein, decreased dihydrofolate reductase, and G(1) arrest with decreased numbers of cells in S-phase. p53 induction was accompanied by up to 3-fold decreases in hRFC transcripts transcribed from the upstream hRFC-B promoter and similar losses of hRFC protein and methotrexate uptake capacity. Expression of p15 in an analogous inducible system in K562 cells resulted in a nearly identical decrease of S-phase cells and dihydrofolate reductase without effects on hRFC levels or activity. When the hRFC-B promoter was expressed as full-length and basal promoter-luciferase reporter constructs in K562(pTet-on/p53) cells, induction of p53 with doxycycline resulted in a 3-fold loss of promoter activity, which was reversed by cotransfection with a trans-dominant-negative p53. These studies show that wild type p53 acts as a repressor of hRFC gene expression, via a mechanism that is independent of its effects on cell cycle progression.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation/physiology , Membrane Proteins , Membrane Transport Proteins , Tumor Suppressor Protein p53/physiology , 5' Untranslated Regions , Humans , Methotrexate/pharmacokinetics , Promoter Regions, Genetic , Reduced Folate Carrier Protein , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
4.
J Biol Chem ; 276(9): 6350-8, 2001 Mar 02.
Article in English | MEDLINE | ID: mdl-11078737

ABSTRACT

Our laboratory previously identified two functional promoters (designated A and B) for the human reduced folate carrier (hRFC) gene that result in hRFC transcripts with differing 5'-untranslated regions. By transiently transfecting HT1080 and HepG2 cells with a series of 5' and 3' deletions in the hRFC-B and -A promoters, the minimal promoters were localized within 46 and 47 base pairs, respectively. Gel mobility shift assays with the hRFC-B basal promoter region revealed specific DNA-protein complexes involving a highly conserved GC-box and Sp1 or Sp3. In Drosophila SL2 cells, both Sp1 and the long Sp3 isoform potently transactivated the hRFC-B basal promoter; however, the short Sp3 isoforms were transcriptionally inert and resulted in a potent inhibition of Sp1 transactivation. For the hRFC-A basal promoter, a CRE/AP-1-like element was bound by the bZip superfamily of DNA-binding proteins. Cell-specific DNA-protein complexes were identified for hRFC-A (CREB-1 and c-Jun in HT1080 cells; CREB-1 and ATF-1 in HepG2 cells). When the GC-box and CRE/AP-1-like elements were mutated, a 60--90% decrease in promoter activity was observed in both cell lines. These results identify the critical regulatory regions for the hRFC basal promoters and stress the functional importance of the Sp and bZip families of transcription factors in regulating hRFC expression.


Subject(s)
Carrier Proteins/genetics , Cyclic AMP/pharmacology , Gene Expression Regulation , Membrane Proteins , Membrane Transport Proteins , Promoter Regions, Genetic , Response Elements , Animals , Base Sequence , DNA-Binding Proteins/physiology , Drosophila , Humans , Molecular Sequence Data , Organ Specificity , Reduced Folate Carrier Protein , Sp1 Transcription Factor/physiology , Sp3 Transcription Factor , Transcription Factors/metabolism , Transcription Factors/physiology , Transcriptional Activation , Transfection
5.
Toxicol Appl Pharmacol ; 168(3): 216-24, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11042094

ABSTRACT

The flavin-containing monooxygenases (FMOs) are a family of xenobiotic-metabolizing enzymes that are expressed in a species- and tissue-specific manner. FMO2 expression has been observed in pulmonary tissue from several species, but not human. Two human FMO2 point mutations have been reported: a cytosine to thymidine transition at position 1414 resulting in a premature stop codon and a thymidine insertion at position 1589 resulting in a frameshift. To define the frequency of these sequence variations and explore their significance, unrelated African-American, Caucasian, and Korean individuals were genotyped. In the African-American population tested (n = 180), the 1414C allele occurred at a 13% frequency; however, all of the tested Caucasians (n = 52) and Koreans (n = 100) were homozygous for the 1414T allele. The T1589 allele occurred at frequencies of 6.9 and 13.0% in African-Americans (n = 175) and Caucasians (n = 23), respectively, and appears to segregate with the 1414T allele. Thus, it would have no further impact on FMO2 activity. Western blot analysis of pulmonary microsomes failed to detect immunoreactive protein in 1414T homozygotes. A heterozygotic individual did exhibit a single band of the expected size, but no detectable FMO activity in the corresponding lung microsomes. Sequence analysis, however, was consistent with the 1414C allele encoding an active FMO2 enzyme. FMO2 mRNA expression was observed in most individuals, but failed to correlate with genotype or protein expression. In summary, functional FMO2 is expressed in only a small percentage of the overall population. However, in certain ethnic groups, active pulmonary FMO2 enzyme will be present in a significant number of individuals.


Subject(s)
Black People/genetics , Oxygenases/genetics , Polymorphism, Genetic/genetics , Alleles , Blotting, Western , Genotype , Humans , Oligonucleotides/analysis , Oligonucleotides/genetics , Oxygenases/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...