Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Ann Oncol ; 28(11): 2725-2732, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28945836

ABSTRACT

BACKGROUND: Thymidylate synthase (TS) has a predictive role in pemetrexed treatment of mesothelioma; however, additional chemoresistance mechanisms are poorly understood. Here, we explored the role of the reduced-folate carrier (RFC/SLC19A1) and proton-coupled folate transporter (PCFT/SLC46A1) in antifolate resistance in mesothelioma. PATIENTS AND METHODS: PCFT, RFC and TS RNA and PCFT protein levels were determined by quantitative RT-PCR of frozen tissues and immunohistochemistry of tissue-microarrays, respectively, in two cohorts of pemetrexed-treated patients. Data were analyzed by t-test, Fisher's/log-rank test and Cox proportional models. The contribution of PCFT expression and PCFT-promoter methylation to pemetrexed activity were evaluated in mesothelioma cells and spheroids, through 5-aza-2'-deoxycytidine-mediated demethylation and siRNA-knockdown. RESULTS: Pemetrexed-treated patients with low PCFT had significantly lower rates of disease control, and shorter overall survival (OS), in both the test (N = 73, 11.3 versus 20.1 months, P = 0.01) and validation (N = 51, 12.6 versus 30.3 months, P = 0.02) cohorts. Multivariate analysis confirmed PCFT-independent prognostic role. Low-PCFT protein levels were also associated with shorter OS. Patients with both low-PCFT and high-TS levels had the worst prognosis (OS, 5.5 months), whereas associations were neither found for RFC nor in pemetrexed-untreated patients. PCFT silencing reduced pemetrexed sensitivity, whereas 5-aza-2'-deoxycytidine overcame resistance. CONCLUSIONS: These findings identify for the first time PCFT as a novel mesothelioma prognostic biomarker, prompting prospective trials for its validation. Moreover, preclinical data suggest that targeting PCFT-promoter methylation might eradicate pemetrexed-resistant cells characterized by low-PCFT expression.


Subject(s)
Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm , Mesothelioma/pathology , Pemetrexed/therapeutic use , Pleural Neoplasms/pathology , Proton-Coupled Folate Transporter/metabolism , Reduced Folate Carrier Protein/metabolism , Adult , Aged , Aged, 80 and over , Cell Proliferation/drug effects , Female , Folic Acid Antagonists/therapeutic use , Follow-Up Studies , Humans , Immunoenzyme Techniques , Male , Mesothelioma/drug therapy , Mesothelioma/metabolism , Middle Aged , Pleural Neoplasms/drug therapy , Pleural Neoplasms/metabolism , Prognosis , Survival Rate , Thymidylate Synthase/metabolism , Tumor Cells, Cultured
2.
Leukemia ; 23(8): 1417-25, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19340001

ABSTRACT

We explored the impact of mutations in the NOTCH1, FBW7 and PTEN genes on prognosis and downstream signaling in a well-defined cohort of 47 patients with pediatric T-cell acute lymphoblastic leukemia (T-ALL). In T-ALL lymphoblasts, we identified high-frequency mutations in NOTCH1 (n=16), FBW7 (n=5) and PTEN (n=26). NOTCH1 mutations resulted in 1.3- to 3.3-fold increased transactivation of an HES1 reporter construct over wild-type NOTCH1; mutant FBW7 resulted in further augmentation of reporter gene activity. NOTCH1 and FBW7 mutations were accompanied by increased median transcripts for NOTCH1 target genes (HES1, DELTEX1 and cMYC). However, none of these mutations were associated with treatment outcome. Elevated HES1, DELTEX1 and cMYC transcripts were associated with significant increases in transcript levels of several chemotherapy relevant genes, including MDR1, ABCC5, reduced folate carrier, asparagine synthetase, thiopurine methyltransferase, BCL2 and dihydrofolate reductase. PTEN transcripts positively correlated with HES1 and cMYC transcript levels. Our results suggest that (1) multiple factors should be considered with attempting to identify molecular-based prognostic factors for pediatric T-ALL, and (2) depending on the NOTCH1 signaling status, modifications in the types or dosing of standard chemotherapy drugs for T-ALL, or combinations of agents capable of targeting NOTCH1, AKT and/or mTOR with standard chemotherapy agents may be warranted.


Subject(s)
Cell Cycle Proteins/genetics , F-Box Proteins/genetics , Gene Expression Regulation, Leukemic/genetics , Mutation , Neoplasm Proteins/genetics , PTEN Phosphohydrolase/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptor, Notch1/genetics , Ubiquitin-Protein Ligases/genetics , Adolescent , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle Proteins/physiology , Child , Child, Preschool , Cohort Studies , DNA Mutational Analysis , DNA, Neoplasm/genetics , F-Box Proteins/physiology , F-Box-WD Repeat-Containing Protein 7 , Female , Genes, Reporter , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Infant , Male , Neoplasm Proteins/physiology , PTEN Phosphohydrolase/physiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prognosis , Receptor, Notch1/physiology , Signal Transduction/genetics , Transcription Factor HES-1 , Treatment Outcome , Ubiquitin-Protein Ligases/physiology , Young Adult
3.
Oncogene ; 27(36): 4933-42, 2008 Aug 21.
Article in English | MEDLINE | ID: mdl-18469864

ABSTRACT

t(8;21)(q22;q22) results in the AML1-ETO (A1E) fusion gene and is a common cytogenetic abnormality in acute myeloid leukemia (AML). Although insertions at the breakpoint region of the A1E fusion transcripts have been reported, additional structural alterations are largely uncharacterized. By RT-PCR amplifications and DNA sequencing, numerous in-frame and out-of-frame AML1b-ETO and AML1c-ETO transcripts were identified in 13 pediatric t(8;21) AMLs, likely resulting from alternate splicing, internal deletions and/or breakpoint region insertions involving both the AML1 (RUNX1) and ETO regions. The in-frame A1E fusion transcript forms represented minor forms. These structure alterations were found in AML1c-ETO but not AML1b-ETO transcripts in two adult t(8;21) AMLs. Although no analogous alterations were detected in native AML1b transcripts, identical alterations in native ETO transcripts were identified. When transfected into HeLa cells, only AML1b, and not the in-frame A1E forms, transactivated the GM-CSF promoter. In co-transfection experiments, the effects of A1E proteins on GM-CSF transactivation by AML1b ranged from repressive to activating. Our results demonstrate a remarkable and unprecedented heterogeneity in A1E fusion transcripts in t(8;21) myeloblasts and suggest that synthesis of alternate A1E transcript and protein forms can significantly impact the regulation of AML1 responsive genes.


Subject(s)
Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Core Binding Factor Alpha 2 Subunit/genetics , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , RNA, Messenger/genetics , Translocation, Genetic , Alternative Splicing , Base Sequence , DNA Primers , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Promoter Regions, Genetic , RUNX1 Translocation Partner 1 Protein , Reverse Transcriptase Polymerase Chain Reaction
4.
Leukemia ; 22(3): 521-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18094719

ABSTRACT

Acute myeloid leukemia (AML) in Down syndrome (DS) children has several unique features including a predominance of the acute megakaryocytic leukemia (AMkL) phenotype, higher event-free survivals compared to non-DS children using cytosine arabinoside (ara-C)/anthracycline-based protocols and a uniform presence of somatic mutations in the X-linked transcription factor gene, GATA1. Several chromosome 21-localized transcription factor oncogenes including ETS2 may contribute to the unique features of DS AMkL. ETS2 transcripts measured by real-time RT-PCR were 1.8- and 4.1-fold, respectively, higher in DS and non-DS megakaryoblasts than those in non-DS myeloblasts. In a doxycycline-inducible erythroleukemia cell line, K562pTet-on/ETS2, induction of ETS2 resulted in an erythroid to megakaryocytic phenotypic switch independent of GATA1 levels. Microarray analysis of doxycycline-induced and doxycycline-uninduced cells revealed an upregulation by ETS2 of cytokines (for example, interleukin 1 and CSF2) and transcription factors (for example, TAL1), which are key regulators of megakaryocytic differentiation. In the K562pTet-on/ETS2 cells, ETS2 induction conferred differences in sensitivities to ara-C and daunorubicin, depending on GATA1 levels. These results suggest that ETS2 expression is linked to the biology of AMkL in both DS and non-DS children, and that ETS2 acts by regulating expression of hematopoietic lineage and transcription factor genes involved in erythropoiesis and megakaryopoiesis, and in chemotherapy sensitivities.


Subject(s)
Gene Expression Regulation, Leukemic/physiology , Leukemia, Myeloid/etiology , Neoplasm Proteins/physiology , Proto-Oncogene Protein c-ets-2/physiology , Acute Disease , Cell Differentiation/genetics , Child , Chromosomes, Human, Pair 21/genetics , Cytarabine/pharmacology , Daunorubicin/pharmacology , Down Syndrome/complications , Drug Resistance, Neoplasm/genetics , Erythroid Precursor Cells/metabolism , Erythropoiesis/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/physiology , Gene Dosage , Gene Expression Regulation, Leukemic/genetics , Genetic Predisposition to Disease , Humans , K562 Cells/drug effects , K562 Cells/metabolism , Leukemia, Megakaryoblastic, Acute/etiology , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/genetics , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Proto-Oncogene Mas , Thrombopoiesis/genetics
5.
Leukemia ; 20(3): 417-25, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16437149

ABSTRACT

Hemgn (a gene symbol for hemogen in mouse, EDAG in human and RP59 in rat) encodes a nuclear protein that is highly expressed in hematopoietic tissues and acute leukemia. To characterize its regulatory mechanisms, we examined the activities of a Hemgn promoter containing 2975 bp of 5' flanking sequence and 196 bp of 5' untranslated region (5' UTR) sequence both in vitro and in vivo: this promoter is preferentially activated in a hematopoietic cell line, not in nonhematopoietic cell lines, and is sufficient to drive the transcription of a lacZ transgene in hematopoietic tissues in transgenic mice. Mutagenesis analyses showed that the 5' UTR including two highly conserved GATA boxes is critical for the promoter activity. GATA1, not GATA2, binds to the GATA binding sites and transactivates the Hemgn promoter in a dose-dependent manner. Furthermore, the expression of human hemogen (EDAG) transcripts were closely correlated with levels of GATA1 transcripts in primary acute myeloid leukemia specimens. This study suggests that the Hemgn promoter contains critical regulatory elements for its transcription in hematopoietic tissues and Hemgn is a direct target of GATA1 in leukemia cells.


Subject(s)
Bone Marrow Cells/metabolism , GATA1 Transcription Factor/physiology , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic , Transcription, Genetic , 5' Untranslated Regions , Animals , Base Sequence , Cell Line, Tumor , Child , DNA , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Transgenic , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction
7.
J Neural Transm Suppl ; (67): 95-103, 2003.
Article in English | MEDLINE | ID: mdl-15068242

ABSTRACT

Down syndrome (trisomy of chromosome 21) (DS) is the most common genetic cause of mental retardation. In our study we employed immunoblotting to evaluate protein expression of reduced folate carrier (hRFC), encoded by a gene localised on chromosome 21, in fetal DS brain. We observed increased expression of hRFC-immunoreactive band with an apparent MW of approximately 150 kDa, whereas the other bands (MWs approximately 60 and 50 kDa), were comparable to control. In conclusion, we suggest that aberrant hRFC expression may well have a role in the already observed deterioration of folate metabolism in DS. Moreover, no alterations of expression level of p53 and Sp1, supposed to play a role in the regulation of hRFC, suggest that regulation of hRFC expression in fetal life by these proteins is highly unlikely, at least by changes in their protein level.


Subject(s)
Cerebral Cortex/metabolism , Down Syndrome/metabolism , Fetus/metabolism , Folic Acid/metabolism , Gene Expression Regulation, Developmental/physiology , Membrane Transport Proteins/biosynthesis , Brain/metabolism , Down Syndrome/genetics , Female , Humans , Membrane Transport Proteins/genetics , Reduced Folate Carrier Protein , Regression Analysis , Statistics, Nonparametric
8.
AAPS PharmSci ; 4(3): E14, 2002.
Article in English | MEDLINE | ID: mdl-12423063

ABSTRACT

Nucleotide efflux (especially cyclic nucleotides) from a variety of mammalian tissues, bacteria, and lower eukaryotes has been studied for several decades. However, the molecular identity of these nucleotide efflux transporters remained elusive, despite extensive knowledge of their kinetic properties and inhibitor profiles. Identification of the subfamily of adenosine triphosphate (ATP) binding cassette transporters, multidrug resistance protein (MRP) subfamily, permitted rapid advances because some recently identified MRP family members transport modified nucleotide analogs (ie, chemotherapeutic agents). We first identified, MRP4, based on its ability to efflux antiretroviral compounds, such as azidothymidine monophosphate (AZT-MP) and 9-(2-phosphonyl methoxyethyl) adenine (PMEA), in drug-resistant and also in transfected cell lines. MRP5, a close structural homologue of MRP4 also transported PMEA. MRP4 and MRP5 confer resistance to cytotoxic thiopurine nucleotides, and we demonstrate MRP4 expression varies among acute lymphoblastic leukemias, suggesting this as a factor in response to chemotherapy with these agents. The ability of MRP4 and MRP5 to transport 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP) suggests they may play a biological role in cellular signaling by these nucleotides. Finally, we propose that MRP4 may also play a role in hepatic bile acid homeostasis because loss of the main bile acid efflux transporter, sister of P-glycoprotein (SPGP) aka bile-salt export pump (BSEP), leads to a strong compensatory upregulation in MRP4 expression. Cumulatively, these studies reveal that the ATP-binding cassette (ABC) transporters MRP4 and MRP5 have a unique role in biology and in chemotherapeutic response.


Subject(s)
Drug Therapy , Multidrug Resistance-Associated Proteins/physiology , Animals , Biological Transport, Active/physiology , Drug Resistance, Neoplasm/physiology , Humans , Multidrug Resistance-Associated Proteins/genetics , Nucleotides, Cyclic/metabolism , T-Lymphocytes/chemistry , T-Lymphocytes/metabolism
9.
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
12.
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
13.
J Biol Chem ; 276(47): 43570-9, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11562358

ABSTRACT

Cystathionine beta-synthase (CBS) catalyzes the condensation of serine and homocysteine to form cystathionine, an intermediate step in the synthesis of cysteine. We previously characterized the CBS -1b minimal promoter (-3792 to -3667) and found that Sp1/Sp3, nuclear factor Y, and USF-1 were involved in the regulation of basal promoter activity (Ge, Y., Konrad, M. A., Matherly, L. H., Taub, J. W. (2001) Biochem. J. 357, 97-105). In this study, the critical cis-elements and transcription factors in the CBS -1b upstream region (-4046 to -3792) were examined in HT1080 and HepG2 cells, which differ approximately 10-fold in levels of CBS transcripts transcribed from the CBS -1b promoter. In DNase I footprint and gel shift analyses and transient transfections of mutant CBS -1b promoter constructs into HT1080 and HepG2 cells, transcriptionally important roles for Sp1/Sp3 binding to three GC boxes and one GT box and for binding of myeloid zinc finger 1-like proteins to two myeloid zinc finger 1 elements were indicated. In gel shift assays, very low levels of Sp1/Sp3 DNA-protein complexes were detected in HT1080 cells compared with HepG2 cells despite comparable levels of nuclear factor Y and USF-1 binding and similar levels of Sp1 and Sp3 proteins on Western blots. Mixing of HT1080 and HepG2 nuclear extracts resulted in no difference in total Sp factor binding in gel shift assays, thus excluding a role for an unknown activator or inhibitor in the disparate Sp1/Sp3 binding between the lines. Increased Sp1/Sp3 binding in gel shift assays was observed upon treatment of HT1080 nuclear extracts with protein kinase A, and decreased Sp1/Sp3 binding resulted from treatment of HepG2 nuclear extracts with calf alkaline phosphatase, suggesting a role for changes in Sp1/Sp3 phosphorylation in transcription factor binding and transactivation of the CBS -1b promoter. Characterization of CBS promoter structure and function should clarify the molecular bases for variations in CBS gene expression in genetic diseases and the relationship between CBS and Down syndrome.


Subject(s)
Cystathionine beta-Synthase/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic/physiology , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Transcription, Genetic/physiology , Base Sequence , Cell Line , DNA , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sp3 Transcription Factor , Transfection
14.
Biochem J ; 358(Pt 2): 511-6, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11513752

ABSTRACT

The human reduced folate carrier (hRFC) is an integral membrane protein that mediates cellular uptake of reduced folates and antifolates. hRFC contains several highly conserved charged residues predicted to lie in the transmembrane domains (TMDs). To explore the possible roles of the conserved arginine-133, located in TMD 4, in hRFC structure and function, this residue was systematically mutagenized to histidine, leucine, lysine and glutamate. When transfected into transport-impaired K562 cells, the mutant hRFC constructs were expressed at high levels; however, only lysine-133 hRFC was able to transport methotrexate and (6S)-5-formyl tetrahydrofolate. Substitution of aspartate-453 (in hRFC TMD 12) by valine largely preserved transport activity for both substrates. Although mutagenesis of aspartate-88 (in TMD 2) to leucine completely abolished transport activity in transfected cells, substitution with a glutamate preserved low levels ( approximately 12%) of transport. To assess the possibility that arginine-133 and aspartate-88 may form a charge-pair to stabilize hRFC tertiary structure, both charges were neutralized (by substituting leucine and valine, respectively) in the same construct. In contrast to the singly mutated hRFCs, the double mutant exhibited high levels of transport with both methotrexate and 5-formyl tetrahydrofolate. These results strongly suggest that arginine-133 and aspartate-88 form a charge-pair and that TMD 4 lies next to TMD 2 in the hRFC tertiary structure.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/physiology , Membrane Transport Proteins , Amino Acid Sequence , Animals , Arginine/physiology , Aspartic Acid/physiology , Biological Transport , Carrier Proteins/genetics , Cricetinae , Humans , K562 Cells , Leucovorin/metabolism , Methotrexate/metabolism , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Rats , Reduced Folate Carrier Protein , Sequence Homology, Amino Acid , Structure-Activity Relationship , Transfection
15.
Article in English | MEDLINE | ID: mdl-11525381

ABSTRACT

The natural folates are water-soluble members of the B class of vitamins that are essential for cell proliferation and tissue regeneration. Since mammalian cells cannot synthesize folates de novo, tightly regulated and sophisticated cellular uptake processes have evolved to sustain sufficient levels of intracellular tetrahydrofolate cofactors to support the biosynthesis of purines, pyrimidines, serine, and methione. Membrane transport is also a critical determinant of the antitumor activity of antifolate therapeutics (methotrexate, Tomudex) used in cancer chemotherapy, and impaired uptake of antifolates is a frequent mode of drug resistance. The reduced folate carrier is the major transport system for folates and classical antifolates in mammalian cells and tissues. This review summarizes the remarkable advances in the cellular and molecular biology of the human reduced folate carrier over the past decade, relating to its molecular structure and transport function, mechanisms of transcriptional and posttranscriptional regulation, and its critical role in antifolate response and resistance. Many key in vitro findings have now begun to be extended to studies of reduced folate carrier levels and function in patient specimens, paving the way for translating basic laboratory studies in cultured cells to improvements in human health and treatment of disease. The results of research into the human reduced folate carrier should clarify the roles of changes in expression and function of this system that accompany nutritional folate deficiency and human disease, and may lead to improved therapeutic strategies for enhancing drug response and circumventing resistance in cancer patients undergoing chemotherapy with antifolates.


Subject(s)
Carrier Proteins/metabolism , Membrane Transport Proteins , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , DNA, Complementary , Drug Resistance, Neoplasm , Folic Acid/metabolism , Humans , Methotrexate/pharmacology , RNA Processing, Post-Transcriptional , Reduced Folate Carrier Protein , Transcription, Genetic
16.
Br J Haematol ; 113(3): 746-56, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11380466

ABSTRACT

The downstream effects of p15 and p16 gene deletions and loss of transcripts on dihydrofolate reductase (DHFR) were examined in 63 B-precursor (BP) acute lymphoblastic leukaemia (ALL) samples. p15 and/or p16 gene deletions were seen in 6% and 8%, respectively, of BP-ALL samples; however, losses of p15 and/or p16 transcripts were seen in 26 out of 63 (41%) samples. Loss of p15 transcripts (36.5%) exceeded that for p16 (17.5%). For the 26 BP-ALLs that lacked p15 and/or p16 transcripts, only six (23%) exhibited low levels of DHFR by flow cytometry assay with Pt430, a fluorescent anti-folate. Conversely, 18 out of 37 (49%) BP-ALL samples with intact p15 and/or p16 genes and transcripts showed low levels of DHFR (P = 0.04). In p15- and p16-null K562 cells transfected with a tetracycline-inducible p15 cDNA construct, induction of p15 transcripts and protein was accompanied by decreased growth rates, decreased S-phase fraction, decreased retinoblastoma protein phosphorylation, and markedly reduced levels of DHFR transcripts and protein. Collectively, our results suggest that losses of p15 and/or p16 gene expression result in elevated levels of DHFR in BP-ALL in children. However, additional downstream factors undoubtedly also contribute to elevated levels of this enzyme target.


Subject(s)
Burkitt Lymphoma/genetics , Cell Cycle Proteins , Gene Deletion , Genes, p16 , Tetrahydrofolate Dehydrogenase/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins , Adolescent , Blotting, Southern , Burkitt Lymphoma/enzymology , Case-Control Studies , Cell Cycle , Child , Child, Preschool , Confidence Intervals , Cyclin-Dependent Kinase Inhibitor p15 , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Female , Flow Cytometry , Gene Expression/drug effects , Humans , Infant , K562 Cells , Logistic Models , Male , Odds Ratio , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
17.
Biochem J ; 357(Pt 1): 97-105, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11415440

ABSTRACT

Cystathionine beta-synthase (CBS) catalyses the condensation of serine and homocysteine to form cystathionine, an intermediate step in the synthesis of cysteine. Human CBS encodes five distinct 5' non-coding exons, the most frequent termed CBS -1a and CBS -1b, each transcribed from its own unique GC-rich TATA-less promoter. The minimal transcriptional region (-3792 to -3667) of the CBS -1b promoter was defined by 5'- and 3'-deletions, and transient transfections of reporter gene constructs in HepG2 cells, characterized by CBS transcription exclusively from the -1b promoter. Included in this 125 bp region are 3 GC-boxes (termed GC-a, GC-b and GC-c), an inverted CAAT-box and an E-box. By gel-shift and supershift assays, binding of specificity protein (Sp)1 and Sp3 to the GC-box elements, upstream stimulatory factor 1 (USF-1) to the E-box, and both nuclear factor (NF)-Y and an NF-1-like factor to the CAAT box could be demonstrated. By transient trans fections and reporter gene assays in HepG2 and Drosophila SL2 cells, a functional interplay was indicated between NF-Y binding to the CAAT-box, or between USF-1 binding to the E-box, and Sp1/Sp3 binding to the GC-box elements. In SL2 cells, NF-Y and Sp1/Sp3 were synergistic. Furthermore, both Sp1 and the long Sp3 isoform transactivated the CBS -1b minimal promoter; however, the short Sp3 isoforms were potent repressors. These results may explain the cell- or tissue-specific regulation of CBS transcription, and clarify the bases for alterations in CBS gene expression in human disease and Down's syndrome.


Subject(s)
CCAAT-Binding Factor/metabolism , Cystathionine beta-Synthase/genetics , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Transcriptional Activation , Base Sequence , Binding Sites , Exons , Gene Expression Regulation, Enzymologic , Humans , Introns , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Recombinant Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured
18.
Biochem Pharmacol ; 61(6): 665-75, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11266651

ABSTRACT

Severe impairment of methotrexate membrane transport in methotrexate-resistant K562 (K500E) cells was characterized by a nearly complete loss of reduced folate carrier (RFC) transcripts and RFC protein. As determined by 5'-rapid amplification of cDNA ends (5'-RACE), approximately 93% of the RFC transcripts in wild-type cells contained the KS43 5'-untranslated region transcribed from the RFC-B promoter. KS43 transcripts decreased > 90% in K500E cells. The basal and full-length RFC-B promoters were more active (3- and 2-fold, respectively) in directing transcription of a luciferase reporter gene in K500E than in wild-type cells. Treatment with a demethylating agent, 5-aza-2'-deoxycytidine, or with a histone deacetylase inhibitor, trichostatin A, did not increase the levels of RFC transcripts in K500E cells. No differences in RFC gene structure were detected between the lines on Southern blots; however, the RFC signals were decreased approximately 60% in K500E cells. DNA sequences were identical between the lines for the RFC coding region and the two 5'-non-coding exons and their respective promoters. Spectral karyotype analysis and fluorescence in situ hybridization in wild-type cells showed two normal chromosome 21 copies and one or two marker chromosomes, each with an RFC signal. In K500E cells, the RFC gene locus was no longer localized to a normal chromosome 21 (at 21q22.2), and a single RFC signal was associated with a small metacentric chromosome, characterized by a 21/22 translocation. Our results suggest that loss of RFC transcripts in K500E cells is unrelated to changes in the levels of critical transcription factors, or to differences in the extent of RFC promoter methylation or core histone deacetylation. Rather, this phenotype is due to the loss of one or more RFC alleles, and to a translocation of the remaining RFC allele with the formation of a 21/22 fusion chromosome.


Subject(s)
Azacitidine/analogs & derivatives , Carrier Proteins/genetics , Gene Deletion , Gene Expression Regulation, Neoplastic , Leukemia, Erythroblastic, Acute/genetics , Membrane Proteins , Membrane Transport Proteins , Translocation, Genetic , 5' Untranslated Regions/genetics , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Biological Transport , Carrier Proteins/biosynthesis , DNA Methylation , Decitabine , Drug Resistance, Neoplasm/genetics , Genes, Reporter , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , In Situ Hybridization, Fluorescence , K562 Cells , Karyotyping , Methotrexate/pharmacokinetics , Methotrexate/pharmacology , Promoter Regions, Genetic/physiology , RNA, Messenger/biosynthesis , Reduced Folate Carrier Protein , Reverse Transcriptase Polymerase Chain Reaction
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...