Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
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.
Leukemia ; 22(9): 1692-7, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18548099

ABSTRACT

Recent studies with very small numbers of patients showed that in some cases of childhood acute lymphoblastic leukemia (ALL), preleukemic cells are detectable on Guthrie cards that were used for newborn screening. We present here the largest series of ALL patients (n=32) in whom Guthrie cards were analyzed for the presence of preleukemic cells. Rearranged immunoglobulin heavy-chain genes were used as a marker for leukemic clones. We combined our set of patients with 17 previously published cases. Preleukemic cells were detected in 31 of all 49 patients (63%). Positive screening cards were not associated with patient's age at diagnosis but were almost always found in patients with hyperdiploidy (10/11; 91%; P=0.04). High birth weight is an established risk factor for childhood ALL. Positive screening cards were strongly associated with low birth weight (P=0.01). In conclusion, the majority of childhood B-precursor ALL arise prior to birth. In the search for causes of childhood leukemia we should concentrate on prenatal factors as well as postnatal factors. Our results suggest that autologous cord bloods could be a poor choice as the source of stem cells for transplantation in leukemia, which may contain preleukemic cells. Pending the development of suitable methods, childhood leukemia is a potentially screenable disease.


Subject(s)
Aneuploidy , Birth Weight , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Preleukemia/pathology , Adolescent , Child , Child, Preschool , Female , Gene Rearrangement , Humans , Immunoglobulin Heavy Chains , Infant , Infant, Newborn , Male , Neonatal Screening , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/embryology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Retrospective Studies
4.
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
5.
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
6.
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
8.
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
11.
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
12.
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
13.
J Pediatr Hematol Oncol ; 23(6): 364-7, 2001.
Article in English | MEDLINE | ID: mdl-11563771

ABSTRACT

Infantile or congenital cases of thrombotic microangiopathy have been reported that were familial and characterized by ongoing microangiopathic hemolysis and thrombocytopenia in the absence of regular fresh-frozen plasma transfusions. The authors describe a child with congenital microangiopathic hemolytic anemia and thrombocytopenia (CMHAT) who has received regular fresh-frozen plasma transfusions since infancy and has never had thrombotic complications. von Willebrand factor (vWF)-cleaving protease activity was studied in the patient's pretransfusion and posttransfusion plasma samples as well as in her parents' plasma. The effects of the patient's and a control subject's plasma on human microvascular endothelial cells were also investigated. Unusually large vWF multimers were present in the patient's plasma both before transfusion (thrombocytopenic) and after transfusion. Unlike cases of chronic relapsing thrombotic thrombocytopenic purpura, vWF-cleaving protease activity was present and treatment of cultured human endothelial cells with the patient's plasma did not induce apoptosis. These findings suggest that the patient with CMHAT may represent a different group in the broad spectrum of thrombotic microangiopathies.


Subject(s)
Anemia, Hemolytic, Congenital/blood , Metalloendopeptidases/blood , Thrombocytopenia/blood , von Willebrand Factor/metabolism , ADAM Proteins , ADAMTS13 Protein , Anemia, Hemolytic, Congenital/complications , Anemia, Hemolytic, Congenital/enzymology , Apoptosis , Blood Transfusion , Child, Preschool , Endothelium, Vascular/pathology , Female , Humans , Infant , Thrombocytopenia/complications , Thrombocytopenia/congenital , Thrombocytopenia/enzymology
14.
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
15.
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
17.
J Pediatr Hematol Oncol ; 22(6): 524-6, 2000.
Article in English | MEDLINE | ID: mdl-11132221

ABSTRACT

Spinal cord compression secondary to metastases is an infrequent complication of childhood cancer. We describe an infant with hepatoblastoma in whom cord compression developed because of extensive epidural metastases during treatment. This is a hitherto undescribed metastatic site for hepatoblastoma.


Subject(s)
Hepatoblastoma/secondary , Liver Neoplasms/diagnosis , Spinal Cord Compression/etiology , Spinal Neoplasms/secondary , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cisplatin/therapeutic use , Embolization, Therapeutic , Fatal Outcome , Fluorouracil/administration & dosage , Hepatoblastoma/diagnosis , Hepatoblastoma/drug therapy , Humans , Infant , Liver Neoplasms/drug therapy , Magnetic Resonance Imaging , Male , Spinal Cord Compression/therapy , Spinal Neoplasms/diagnosis , Spinal Neoplasms/drug therapy , Vincristine/therapeutic use
18.
Cancer Res ; 60(22): 6421-6, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11103808

ABSTRACT

The significantly higher event-free survival rates of Down syndrome (DS) children with acute myeloid leukemia compared with non-DS children is linked to increased sensitivity of DS myeloblasts to 1-beta-D-arabinofuranosylcytosine (ara-C) and the enhanced metabolism of ara-C to ara-C triphosphate (J. W. Taub et al., Blood, 87: 3395-3403, 1996). The cystathionine-beta-synthase (CBS) gene (localized to chromosome 21q22.3) may have downstream effects on reduced folate and S-adenosylmethionine pathways; ara-C metabolism and folate pools are linked by the known synergistic effect of sequential methotrexate and ara-C therapy. We have shown that relative CBS transcripts were significantly higher in DS compared with non-DS myeloblasts, and CBS transcript levels correlated with in vitro ara-C sensitivity (J. W. Taub et al., Blood, 94: 1393-1400, 1999). A leukemia cell line model to study the relationship of the CBS gene and ara-C metabolism/sensitivity was developed by transfecting CBS-null CCRF-CEM cells with the CBS cDNA. CBS-transfected cells were a median 15-fold more sensitive in vitro to ara-C compared with wild-type cells and generated 8.5-fold higher [3H]ara-C triphosphate levels after in vitro incubation with [3H]ara-C. Severe combined immunodeficient mice implanted with CBS-transfected CEM cells demonstrated greater responsiveness to therapy, reflected in significantly prolonged survivals after ara-C administration compared with mice implanted with wild-type cells and treated with the same dosage schedule. The transfected cells also demonstrated increased in vitro and in vivo sensitivity to gemcitabine. Deoxycytidine kinase (dCK) activity was approximately 22-fold higher in transfected CEM cells compared with wild-type cells. However, levels of dCK transcripts on Northern blots and protein levels on Western blots were nearly identical between CBS-transfected and wild-type cells. Collectively, these results suggest a posttranscriptional regulation of dCK in CBS-overexpressing cells that contributes to increased ara-C phosphorylation and drug activity. Further elucidating the mechanisms of increased sensitivity of DS cells to ara-C related to the CBS gene may lead to the application of these novel approaches to acute myeloid leukemia therapy for non-DS patients.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cystathionine beta-Synthase/genetics , Cytarabine/pharmacology , DNA, Complementary/genetics , Down Syndrome/complications , Leukemia, Experimental/enzymology , Animals , Antimetabolites, Antineoplastic/metabolism , Chromosomes, Human, Pair 21/genetics , Cystathionine beta-Synthase/biosynthesis , Cystathionine beta-Synthase/metabolism , Cytarabine/metabolism , Deoxycytidine Kinase/metabolism , Down Syndrome/enzymology , Down Syndrome/genetics , Female , Gene Expression , Humans , Leukemia, Experimental/drug therapy , Leukemia, Experimental/genetics , Male , Mice , Mice, Inbred ICR , Mice, SCID , Transfection , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Leuk Lymphoma ; 35(1-2): 1-20, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10512159

ABSTRACT

The improved outlook for children diagnosed today with acute lymphoblastic leukemia (ALL) over that 40 years ago is remarkable. With modern therapies and supportive care, complete remissions are achieved in up to 95% of patients and long-term disease-free survival rates approach 80%. Methotrexate is a key component in ALL consolidation and maintenance therapies and is administered intrathecally in the prophylaxis and treatment of central nervous system leukemia. Recent reports have significantly extended the results of preclinical studies of methotrexate response and resistance to patients with ALL. The application of new and sensitive molecular biology techniques makes it possible to study specific chromosomal and genetic alterations [t(12;21), hyperdiploidy, deletions or methylation of p15INK4B and p16INK4A] which potentially contribute to methotrexate response and resistance in childhood ALL. Studies of the relationships between genetic alterations and ALL progression, methotrexate pharmacology, and long term event-free-survivals may lead to the better identification of subgroups of patients who exhibit unique levels of sensitivity or resistance to chemotherapy including methotrexate. Further, by characterizing the roles of translocation-generated fusion genes (TEL-AML 1) and tumor suppressor genes (p15INK4B and p16INK4A) in treatment response, it may be possible to identify new and selective targets and/or treatment strategies for both children and adults with ALL who are refractory to current therapies.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Methotrexate/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Antimetabolites, Antineoplastic/metabolism , Child , Disease Progression , Folic Acid/metabolism , Humans , Methotrexate/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Recurrence , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...