Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Leukemia ; 29(9): 1868-74, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25915824

ABSTRACT

The contribution of microRNAs to lymphoma biology is not fully understood. In particular, it remains untested whether microRNA dysregulation could contribute to the emergence of the aggressive subset of B-cell lymphomas that coexpress MYC and BCL2. Here, we identify microRNA-124 (miR-124) as a negative regulator of MYC and BCL2 expression in B-cell lymphomas. Concordantly, stable or transient ectopic expression of miR-124 suppressed cell proliferation and survival, whereas genetic inhibition of this miRNA enhanced the fitness of these tumors. Mechanistically, the activities of miR-124 towards MYC and BCL2 intersect with both oncogenic and tumor-suppressive pathways. In respect to the former, we show that miR-124 directly targets nuclear factor-κB (NF-κB) p65, and using genetic approaches, we demonstrate that this interaction accounts for the miR-124-mediated suppression of MYC and BCL2. We also characterized miR-124 promoter region and identified a functional p53 binding site. In agreement with this finding, endogenous or ectopic expression of wild-type, but not mutant, p53 increased miR-124 levels and suppressed p65, MYC and BCL2. Our data unveil an miRNA-dependent regulatory circuitry that links p53 to the NF-κB pathway, which when disrupted in B-cell lymphoma may be associated with aberrant coexpression of MYC and BCL2 and poor prognosis.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , MicroRNAs/genetics , NF-kappa B/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , 3' Untranslated Regions , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cloning, Molecular , Gene Expression , Genes, Reporter , Genes, bcl-2 , Genes, myc , Humans , Mutagenesis, Site-Directed , NF-kappa B/genetics , Promoter Regions, Genetic , RNA Interference , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Suppressor Protein p53/genetics
2.
Leukemia ; 29(4): 968-76, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25311243

ABSTRACT

Growing evidence suggests that microRNAs (miRNAs) facilitate the cross-talk between transcriptional modules and signal transduction pathways. MYC and NOTCH1 contribute to the pathogenesis of lymphoid malignancies. NOTCH induces MYC, connecting two signaling programs that enhance oncogenicity. Here we show that this relationship is bidirectional and that MYC, via a miRNA intermediary, modulates NOTCH. MicroRNA-30a (miR-30a), a member of a family of miRNAs that are transcriptionally suppressed by MYC, directly binds to and inhibits NOTCH1 and NOTCH2 expression. Using a murine model and genetically modified human cell lines, we confirmed that miR-30a influences NOTCH expression in a MYC-dependent fashion. In turn, through genetic modulation, we demonstrated that intracellular NOTCH1 and NOTCH2, by inducing MYC, suppressed miR-30a. Conversely, pharmacological inhibition of NOTCH decreased MYC expression and ultimately de-repressed miR-30a. Examination of genetic models of gain and loss of miR-30a in diffuse large B-cell lymphoma (DLBCL) and T-acute lymphoblastic leukemia (T-ALL) cells suggested a tumor-suppressive role for this miRNA. Finally, the activity of the miR-30a-NOTCH-MYC loop was validated in primary DLBCL and T-ALL samples. These data define the presence of a miRNA-mediated regulatory circuitry that may modulate the oncogenic signals originating from NOTCH and MYC.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma, Large B-Cell, Diffuse/genetics , MicroRNAs/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Primary Cell Culture , Proto-Oncogene Proteins c-myc/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
3.
Int J Genomics ; 2013: 465727, 2013.
Article in English | MEDLINE | ID: mdl-24350238

ABSTRACT

Hypothesizing that the Amazonian water system differences would affect the expression of muscle growth-related genes in juvenile tambaqui Colossoma macropomum (Cuvier 1818), this study aimed to analyze the morphometric data and expression of myogenic regulatory factors (MRFs) in the white and red muscle from tambaqui obtained from clear and black Amazonian water systems. All of the MRF transcript levels (myod, myf5, myogenin, and mrf4) were significantly lower in the red muscle from black water fish in comparison to clear water fish. However, in white muscle, only the myod transcript level was significantly decreased in the black water tambaqui. The changes in MRFs gene expression in muscle fibers of tambaqui from black water system provide relevant information about the environmental influence as that of water systems on gene expression of muscle growth related genes in the C. macropomum. Our results showed that the physical and chemical water characteristics change the expression of genes that promote muscle growth, and these results may be also widely applicable to future projects that aim to enhance muscle growth in fish that are of substantial interest to the aquaculture.

4.
Int J Sports Med ; 34(4): 293-301, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23059557

ABSTRACT

The purpose of this study was to test the hypothesis that skeletal muscle adaptations induced by long-term resistance training (RT) are associated with increased myogenic regulatory factors (MRF) and insulin-like growth factor-I (IGF-I) mRNA expression in rats skeletal muscle. Male Wistar rats were divided into 4 groups: 8-week control (C8), 8-week trained (T8), 12-week control (C12) and 12-week trained (T12). Trained rats were submitted to a progressive RT program (4 sets of 10-12 repetitions at 65-75% of the 1RM, 3 day/week), using a squat-training apparatus with electric stimulation. Muscle hypertrophy was determined by measurement of muscle fiber cross-sectional area (CSA) of the muscle fibers, and myogenin, MyoD and IGF-I mRNA expression were measured by RT-qPCR. A hypertrophic stabilization occurred between 8 and 12 weeks of RT (control-relative % area increase, T8: 29% vs. T12: 35%; p>0.05) and was accompanied by the stabilization of myogenin (control-relative % increase, T8: 44.8% vs. T12: 37.7%, p>0.05) and MyoD (control-relative % increase, T8: 22.9% vs. T12: 22.3%, p>0.05) mRNA expression and the return of IGF-I mRNA levels to the baseline (control-relative % increase, T8: 30.1% vs. T12: 1.5%, p<0.05). Moreover, there were significant positive correlations between the muscle fiber CSA and mRNA expression for MyoD (r=0.85, p=0.0001), myogenin (r=0.87, p=0.0001), and IGF-I (r=0.88, p=0.0001). The significant (p<0.05) increase in myogenin, MyoD and IGF-I mRNA expression after 8 weeks was not associated with changes in the fiber-type frequency. In addition, there was a type IIX/D-to-IIA fiber conversion at 12 weeks, even with the stabilization of MyoD and myogenin expression and the return of IGF-I levels to baseline. These results indicate a possible interaction between MRFs and IGF-I in the control of muscle hypertrophy during long-term RT and suggest that these factors are involved more in the regulation of muscle mass than in fiber-type conversion.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/metabolism , MyoD Protein/metabolism , Myogenin/metabolism , Physical Conditioning, Animal/methods , Adaptation, Physiological , Animals , Hypertrophy , Insulin-Like Growth Factor I/genetics , Male , Muscle, Skeletal/pathology , MyoD Protein/genetics , Myogenin/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
7.
J Clin Endocrinol Metab ; 86(6): 2890-4, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11397905

ABSTRACT

Pheochromocytomas are neural crest-derived tumors that occur mostly sporadically, but may also be part of inherited syndromes. The molecular pathogenesis of sporadic pheochromocytomas remains unknown. Recently, the susceptibility gene for familial paraganglioma syndrome, a disorder embryologically related to pheochromocytomas, was characterized and shown to encode the small subunit of succinate dehydrogenase (SDHD), which is part of the mitochondrial complex II. This complex regulates oxygen-sensing signals. Importantly, hypoxic signals also appear to be related to the pathogenesis of pheochromocytomas associated with von Hippel-Lindau syndrome. We sequenced the entire coding region of the SDHD gene in a series of pheochromocytomas. Although we did not find mutations in the gene, we identified a new intronic single nucleotide polymorphism in 15% of the samples (g.97739A-->G). We also confirmed the existence of a sequence highly homologous to the SDHD complementary DNA in chromosome 1p34--36, a region commonly deleted in pheochromocytomas. Full analysis of this sequence revealed a heterozygous single base substitution in 70% of our samples that was also present in the germline. This sequence does not appear to be transcribed and is probably a processed pseudogene. Therefore, despite its chromosomal location, it is unlikely that this sequence is a target of loss of heterozygosity in pheochromocytomas. In conclusion, mutations of the SDHD gene are not a common event in this series of sporadic pheochromocytomas. The existence of SDHD pseudogenes should be considered when analyzing complementary DNA-based samples.


Subject(s)
Adrenal Gland Neoplasms/genetics , Genetic Predisposition to Disease/genetics , Membrane Proteins/genetics , Paraganglioma/genetics , Pheochromocytoma/genetics , Amino Acid Sequence/genetics , Base Sequence/genetics , Chromosomes, Human, Pair 1/genetics , Humans , Molecular Sequence Data , Polymorphism, Genetic/genetics , Pseudogenes/genetics , Succinate Dehydrogenase , Syndrome , Transcription, Genetic
8.
Blood ; 96(13): 4328-34, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11110709

ABSTRACT

Clinical risk factor models such as the International Prognostic Index are used to identify diffuse large B-cell lymphoma (DLB-CL) patients with different risks of death from their diseases. To elucidate the molecular bases for these observed clinical differences in outcome, differential display was used to identify a novel gene, termed BAL (B-aggressive lymphoma), which is expressed at significantly higher levels in fatal high-risk DLB-CLs than in cured low-risk tumors. The major BAL complementary DNA encodes a previously uncharacterized 88-kd nuclear protein with a duplicated N-terminal domain homologous to the nonhistone portion of histone-macroH2A and a C-terminal alpha-helical region with 2 short coiled-coil domains. Of note, the BAL N-terminus and secondary structure resemble those of a recently identified human protein, KIAA1268. In addition, both BAL and KIAA1268 map to chromosome 3q21, further suggesting that these genes belong to a newly identified family. BAL is expressed at increased levels in DLB-CL cell lines with an activated peripheral B cell, rather than a germinal center B cell, phenotype. This observation and the characteristic dissemination of high risk DLB-CLs prompted studies regarding the role of BAL in B-cell migration. In classical transwell assays, stable BAL-overexpressing B-cell lymphoma transfectants had significantly higher rates of migration than vector-only transfectants, indicating that the risk-related BAL gene promotes malignant B-cell migration. (Blood. 2000;96:4328-4334)


Subject(s)
B-Lymphocytes/pathology , Cell Movement/genetics , Genes , Lymphoma, Large B-Cell, Diffuse/genetics , Neoplasm Proteins/genetics , Amino Acid Sequence , B-Lymphocytes/drug effects , Cell Movement/drug effects , Chemokine CXCL12 , Chemokines, CXC/pharmacology , Cloning, Molecular , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/pathology , Molecular Sequence Data , Neoplasm Proteins/physiology , Poly(ADP-ribose) Polymerases , Recombinant Fusion Proteins/physiology , Recombinant Proteins/pharmacology , Risk , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology
9.
Blood ; 94(7): 2403-13, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10498613

ABSTRACT

Protein tyrosine phosphatases (PTP) regulate the proliferation, differentiation, and viability of lymphocytes by modulating their signaling pathways. By using the differential display assay, we have cloned a putative receptor-type PTP, which is predominantly expressed in B-lymphoid tissues (lymph nodes and spleen). This PTP, termed PTPROt (truncated), is a tissue-specific alternatively-spliced form of a human epithelial PTP, PTPRO (PTPU2/GLEPP1). Whereas the epithelial PTPRO includes an approximately 800-amino acid extracellular domain, the major (3 kb) PTPROt cDNA predicts a unique 5' untranslated region and truncated (8 amino acids) extracellular domain with a conserved transmembrane region and single catalytic domain. PTPROt cDNAs encode functional approximately 47-kD and approximately 43-kD PTPs, which are most abundant in normal naive quiescent B cells and decreased or absent in germinal center B cells and germinal center-derived diffuse large B-cell lymphomas. Because PTPROt was predominantly expressed in naive quiescent B cells, the enzyme's effects on cell-cycle progression were examined. When multiple stable PTPROt sense, antisense, and vector only B-cell transfectants were grown in reduced serum and synchronized with nocodazole, PTPROt sense clones exhibited markedly increased G0/G1 arrest. Taken together, these data implicate PTPROt in the growth control of specific B-cell subpopulations.


Subject(s)
Alternative Splicing , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , Cell Cycle/physiology , Protein Tyrosine Phosphatases/genetics , Amino Acid Sequence , Base Sequence , Cell Cycle/drug effects , Cloning, Molecular , Conserved Sequence , DNA, Complementary , G1 Phase , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Humans , Introns , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/genetics , Molecular Sequence Data , Nocodazole/pharmacology , Palatine Tonsil/immunology , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Resting Phase, Cell Cycle , Sequence Deletion , Spleen/immunology , Transfection , Tumor Cells, Cultured
10.
Cancer Genet Cytogenet ; 113(1): 70-2, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10459350

ABSTRACT

We have recently cloned the inv(8)(p11q13) in a patient with acute myeloid leukemia (AML), and demonstrated a fusion between the MOZ and TIF2 genes at 8p11 and 8q13, respectively. We have partially characterized a further case of AML with the same karyotypic abnormality. Rearrangements were detected by Southern blotting with a TIF2 probe that was close to the breakpoint in the original inv(8) case and with a MOZ probe corresponding to the breakpoint cluster region in the t(8;16) (p11;p13). These findings indicate the existence of breakpoint cluster regions within both genes and demonstrate that the MOZ-TIF2 fusion is consistently associated with the inv(8)(p11q13).


Subject(s)
Acetyltransferases/genetics , Chromosome Aberrations/genetics , Chromosome Inversion , Chromosomes, Human, Pair 8 , Leukemia, Myelomonocytic, Acute/genetics , Transcription Factors/genetics , Blotting, Southern , Chromosome Disorders , Histone Acetyltransferases , Humans , Karyotyping , Models, Genetic , Molecular Sequence Data , Nuclear Receptor Coactivator 2
11.
Hum Mol Genet ; 8(2): 185-93, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9931326

ABSTRACT

PTEN is a novel tumour suppressor gene that encodes a dual-specificity phosphatase with homology to adhesion molecules tensin and auxillin. It recently has been suggested that PTEN dephosphorylates phosphatidylinositol 3,4,5-trisphosphate [PtdIns(3, 4,5)P3], which mediates growth factor-induced activation of intracellular signalling, in particular through the serine-threonine kinase Akt, a known cell survival-promoting factor. PTEN has been mapped to 10q23.3, a region disrupted in several human tumours including haematological malignancies. We have analysed PTEN in a series of primary acute leukaemias and non-Hodgkin's lymphomas (NHLs) as well as in cell lines. We have also examined whether a correlation could be found between PTEN and Akt levels in these samples. We show here that the majority of cell lines studied carries PTEN abnormalities. At the structural level, we found mutations and hemizygous deletions in 40% of these cell lines, while a smaller number of primary haematological malignancies, in particular NHLs, carries PTEN mutations. Moreover, one-third of the cell lines had low PTEN transcript levels, and 60% of these samples had low or absent PTEN protein, which could not be attributed to gene silencing by hypermethylation. In addition, we found that PTEN and phosphorylated Akt levels are inversely correlated in the large majority of the examined samples. These findings suggest that PTEN plays a role in the pathogenesis of haematological malignancies and that it might be inactivated through a wider range of mechanisms than initially considered. The finding that PTEN levels inversely correlate with phosphorylated Akt supports the hypothesis that PTEN regulates PtdIns(3,4,5)P3and suggests a role for PTEN in apoptosis.


Subject(s)
Hematologic Neoplasms/genetics , Phosphoric Monoester Hydrolases/genetics , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins , Blotting, Northern , DNA Mutational Analysis , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , HL-60 Cells , Hematologic Neoplasms/pathology , Humans , K562 Cells , Methylation , Mutation , Neoplasm Proteins/analysis , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/metabolism , Proto-Oncogene Proteins c-akt , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion , Transcription, Genetic , Tumor Cells, Cultured
13.
Br J Cancer ; 78(5): 601-5, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9744498

ABSTRACT

Deletions and apparent transcriptional abnormalities of the FHIT gene at 3p14.2 have recently been reported in a wide variety of solid tumours. To determine whether lesions of this gene also occur in leukaemia, we have analysed a total of 97 patients (chronic myeloid leukaemia, CML, in chronic phase or blast crisis, n = 71; de novo acute leukaemia, n = 26) and 16 normal individuals. Intact FHIT transcripts from all cases were amplified using RT-PCR. In addition, smaller size bands that were less intense than the full-length products were amplified from several samples from patients with leukaemia and also from normal leucocytes. Sequencing of the small products revealed that they were derived from FHIT transcripts lacking whole exons. Using single-strand conformation polymorphism analysis, no mutations in the coding sequence were detected in any patient. Furthermore, loss of heterozygosity was not seen in any of 36 informative patients at D3S1300 or D3S1481, markers located within the FHIT locus. We conclude that the FHIT gene and other uncharacterized tumour-suppressor genes at 3p14.2 are unlikely to be involved in the pathogenesis of acute leukaemia or progression of CML from chronic phase to blast crisis. Moreover, low-abundance FHIT transcripts that lack whole exons are not specific to malignant cells and should not be taken as evidence of an abnormality in the absence of demonstrable genomic DNA lesions.


Subject(s)
Acid Anhydride Hydrolases , Blast Crisis/genetics , Chromosomes, Human, Pair 3/genetics , Hematopoietic Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia/genetics , Neoplasm Proteins , Proteins/genetics , Acute Disease , Blast Crisis/pathology , DNA, Neoplasm/analysis , Gene Expression , Humans , Leukemia/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Loss of Heterozygosity , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational
14.
Blood ; 92(5): 1735-42, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9716603

ABSTRACT

The 8p11 myeloproliferative syndrome is a rare, aggressive condition associated with reciprocal translocations of chromosome band 8p11, most commonly the t(8;13)(p11;q12). To identify the genes involved in this translocation, we used fluorescence in situ hybridization (FISH) analysis to show that the chromosome 8 breakpoints fell within YAC 899e2 and that the chromosome 13 breakpoints are clustered in a region flanked by YACs 929f11 and 911h8. FISH using chromosome 13 PAC clones indicated that the t(8;13) is not simply a reciprocal translocation but also involves an inversion of 13q11-12. Exon trapping of a PAC that spanned the chromosome 13 translocation breakpoints led to the identification of a gene, ZNF198, that detected rearranged bands when used as a probe against Southern blots of patient DNA. Conceptual translation of the full-length ZNF198 cDNA sequence predicts a protein of 1377 amino acids that shows significant homology to the DXS6673E/KIAA0385 and KIAA0425 proteins. Alignment of these three proteins revealed a novel, conserved Zn-finger-related motif (MYM domain) of the general form CX2C19-22CX3CX13-19CX2CX19-25FCX3CX3F/Y that is repeated five times in each protein. To identify the translocation partner gene on chromosome 8, 5' and 3' RACE polymerase chain reactions (PCRs) were performed on patient RNA with several combinations of ZNF198 primers. Clones were identified in which the ZNF198 was fused to exon 9 of the fibroblast growth factor receptor-1 (FGFR1), a gene known to map to 8p11. An identical ZNF198-FGFR1 fusion was detected in the three patients with a t(8;13) for whom RNA was available; reciprocal FGFR1-ZNF198 transcripts were not detected. Rearrangements of both ZNF198 and FGFR1 were found in two further patients by Southern blotting. ZNF198-FGFR1 includes the five MYM domains of ZNF198 and the intracellular tyrosine kinase domain of FGFR1. We hypothesize that this fusion leads to constitutive activation of the FGFR1 tyrosine kinase in a manner analogous to the activation of ABL by BCR in chronic myeloid leukemia.


Subject(s)
Carrier Proteins , Chromosomes, Human, Pair 13 , Chromosomes, Human, Pair 8 , DNA-Binding Proteins/genetics , Fibroblast Growth Factor 1/genetics , Myeloproliferative Disorders/genetics , Translocation, Genetic , Amino Acid Sequence , Base Sequence , Chromosomes, Artificial, Yeast , DNA-Binding Proteins/chemistry , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Polymerase Chain Reaction , RNA-Directed DNA Polymerase , Sequence Alignment , Transcription Factors , Zinc Fingers
15.
Leuk Lymphoma ; 29(1-2): 17-26, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9638972

ABSTRACT

Second primary cancers represent an important complication of modern chemotherapy and radiotherapy. Therapy-related (tr) leukemias are among the most common second malignancies in both pediatric and adult populations. Whereas a reasonable amount of data is available regarding the epidemiology, molecular pathogenesis, clinical behavior and response to therapy of second primary acute leukemias, very little is known about therapy-related chronic myeloid leukemia (tr-CML). A better characterization of this entity could increase our understanding about the mechanisms of carcinogenesis, specially the induction of specific genetic abnormalities, e.g., BCR-ABL fusion, following chemotherapy and/or radiotherapy exposure, could facilitate the investigation of the kinetics of the development of CML, and also provide a model to study molecular events that might precede its development. Review of 32 tr-CML cases suggests that there are no clinically appreciable differences between tr-CML and de novo CML cases. Analysis of large epidemiological studies that investigated the risk of second primary leukemias has not shown any clear evidence of a higher risk of CML among individuals who underwent treatment for a primary cancer over the general population. The cancer-predisposing syndromes, the detection of BCR-ABL transcripts in healthy individuals, and the induction in vitro of BCR-ABL fusions by ionizing radiation, are all discussed in the context of tr-CML. Finally, the need for a large epidemiological study to specifically assess the risk of developing second primary CML after chemotherapy and/or radiotherapy is stressed.


Subject(s)
Antineoplastic Agents/adverse effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Leukemia, Radiation-Induced/etiology , Neoplasms, Second Primary/etiology , Humans , Incidence , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/chemically induced , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/epidemiology , Leukemia, Radiation-Induced/epidemiology , Neoplasms, Second Primary/epidemiology , Radiotherapy/adverse effects
16.
Blood ; 91(9): 3127-33, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9558366

ABSTRACT

Chromosomal abnormalities of band 8p11 are associated with a distinct subtype of acute myeloid leukemia with French-American-British M4/5 morphology and prominent erythrophagocytosis by the blast cells. This subtype is usually associated with the t(8;16)(p11;p13), a translocation that has recently been shown to result in a fusion between the MOZ and CBP genes. We have cloned the inv(8)(p11q13), an abnormality associated with the same leukemia phenotype, and found a novel fusion between MOZ and the nuclear receptor transcriptional coactivator TIF2/GRIP-1/NCoA-2. This gene has not previously been implicated in the pathogenesis of leukemia or other malignancies. MOZ-TIF2 retains the histone acetyltransferase homology domains of both proteins and also the CBP binding domain of TIF2. We speculate that the apparently identical leukemia cell phenotype observed in cases with the t(8;16) and the inv(8) arises by recruitment of CBP by MOZ-TIF2, resulting in modulation of the transcriptional activity of target genes by a mechanism involving abnormal histone acetylation.


Subject(s)
Acetyltransferases/genetics , Leukemia, Myeloid, Acute/genetics , Transcription Factors/genetics , Amino Acid Sequence , Base Sequence , Chromatin/metabolism , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 8 , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Histone Acetyltransferases , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Nuclear Proteins/metabolism , Nuclear Receptor Coactivator 2 , Recombinant Fusion Proteins/genetics , Translocation, Genetic
17.
Oncogene ; 16(1): 69-76, 1998 Jan 08.
Article in English | MEDLINE | ID: mdl-9467944

ABSTRACT

The molecular mechanisms leading to Cushing's disease are unclear. Inhibitors of cyclin-cyclin dependent kinase (CDK) complexes are regulators of the cell cycle and may function as tumour suppressor genes, many of which have been involved in the pathogenesis of several human malignancies. A member of this family, the p27/kip1 gene, maps to chromosome 12p13 and encodes an inhibitor of several cyclin-CDK complexes; these control the progression of the cell cycle from G1 to S-phase. Complete lack of p27/kip1 function, as occurs in the p27/kip1 'knockout' mouse, produces a complex phenotype associated with the development of pituitary tumours, specifically those of the intermediate lobe corticotrophs. We therefore investigated whether structural and functional abnormalities of the p27/kip1 gene and loss at the chromosome 12p13 region were present in human corticotrophin (ACTH)-secreting pituitary tumours. We studied 21 pituitary tumours, of which 20 were ACTH-secreting (two of these had biochemical and histological features of 'intermediate-lobe' tumours and one was malignant) while the remaining tumour was a prolactinoma; three ectopic secretors of ACTH (two bronchial and one thymic carcinoid); and a non-secretory thymic carcinoid. The whole coding region of the p27/ kip1 gene was screened for mutations by PCR-SSCP analysis and/or direct sequencing, while tumour mRNA expression was analysed by means of a semi-quantitative duplex PCR. Three polymorphic microsatellite markers of the 12p13 region were used to assess loss of heterozygosity (LOH) in 12 samples. Finally, tumour p27/kip1 protein expression was assessed by immunohistochemistry using a monoclonal antibody in 12 samples suitable for analysis. No sequence abnormalities were found in any of the samples other than a previously-described polymorphism. No LOH was observed in the tumours analysed. p27/kip1 mRNA expression was similar in tumour samples in comparison with normal pituitaries. Seven of the eight corticotroph tumours analysed by immunohistochemistry stained positive for p27/kip1, including the intermediate lobe. The only malignant pituitary tumour in the original series showed an absence of staining for p27/kip1. In addition, the three carcinoid tumours studied were negative on immunohistochemistry. Of a further three malignant pituitary tumours assessed, two (including a prolactinoma) were essentially negative, while the third was moderately positive. We conclude that mutations of the p27/kip1 gene, deletions of the 12p13 area or changes in expression, are not a general feature of corticotroph tumours, even those with intermediate lobe characteristics. However, other mechanisms of p27/kip1 inactivation, such as an abnormality at the post-translational level, may be related to more aggressive histological subtypes of ACTH-secreting and possibly other pituitary tumours.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Carcinoid Tumor/genetics , Cell Cycle Proteins , Microtubule-Associated Proteins/genetics , Mutation , Pituitary Neoplasms/genetics , Tumor Suppressor Proteins , Adult , Aged , Carcinoid Tumor/metabolism , Cushing Syndrome/genetics , Cushing Syndrome/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Female , Humans , Immunohistochemistry , Male , Middle Aged , Pituitary Neoplasms/metabolism
18.
Genes Chromosomes Cancer ; 20(4): 408-11, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9408758

ABSTRACT

We have used Southern blotting and fluorescence in situ hybridization (FISH) to define the breakpoints of a reciprocal translocation, t(10;12)(q24;p13), acquired as a secondary abnormality in a patient with Philadelphia chromosome positive chronic myeloid leukemia (CML) in transformation. A YAC clone that spanned the breakpoint at 12p13 was identified; this YAC included the CDKN1B gene but did not include ETV6. Neither ETV6 nor CDKN1B was rearranged, as determined by FISH and Southern blotting; however, a small deletion encompassing the translocated CDKN1B allele was detected. Analysis of two candidate genes at 10q24, HOX11 and NFKB2 suggested that they are not involved in this translocation. The preliminary mapping of breakpoints in this case demonstrated that they are different from an apparently identical translocation identified previously in a patient with myelodysplastic syndrome. The identification of the split YAC and small deletion should enable a more focused search for a gene or genes that may contribute to progression from chronic phase to blast crisis in CML.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 12/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Repressor Proteins , Translocation, Genetic , Blotting, Southern , Chromosome Aberrations/genetics , Chromosome Mapping , Chromosomes, Artificial, Yeast , Chromosomes, Human, Pair 22/genetics , Chromosomes, Human, Pair 9/genetics , DNA Probes , DNA-Binding Proteins/genetics , Female , Homeodomain Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Karyotyping , NF-kappa B/genetics , NF-kappa B p52 Subunit , Neoplasm Proteins/genetics , Oncogene Proteins/genetics , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-ets , Transcription Factors/genetics , ETS Translocation Variant 6 Protein
19.
Blood ; 90(8): 3130-5, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9376594

ABSTRACT

Two distinct leukemia syndromes are associated with abnormalities of chromosome band 8p11. First, a myeloproliferative disorder with features characteristic of both chronic myeloid leukemia and non-Hodgkin's lymphoma and second, an acute myeloid leukemia (AML) with French-American-British (FAB) M4/5 morphology and prominent erythrophagocytosis. The two syndromes are exemplified by a t(8;13)(p11;q12) and a t(8;16)(p11;p13), respectively, but cytogenetic variants of both have been described. Recently, the t(8;16) has been cloned and shown to fuse the MOZ gene at 8p11 to the CBP gene at 16p13. We have used fluorescence in situ hybridization (FISH), Southern blotting, and reverse transcriptase-polymerase chain reaction (RT-PCR) to refine the 8p11 breakpoint in three cases with t(8;13)(p11;q12) and in a single case of AML-M5 with a clinical picture apparently identical to that found in patients with a t(8;16), but characterized by an inv(8)(p11q13). FISH analysis was performed with several 8p11 CEPH yeast artificial chromosome (YAC) clones. YAC 782H11 was centromeric to the one case with t(8;13) tested, but was telomeric to the inv(8). YAC 847B12 was telomeric to both the t(8;13) and the inv(8), whereas YAC 829D12 was centromeric to the t(8;13), but split by the inv(8). Southern blotting and PCR of YAC 829D12 showed that it contained the MOZ gene. A 900-bp MOZ fragment encompassing the published t(8;16) breakpoint was amplified by PCR from normal peripheral blood leukocyte cDNA and used to probe Southern blots of patient DNA. A rearrangement was detected in the case with inv(8), but not in any of the three cases with t(8;13). Southern blotting with a CBP probe and RT-PCR with MOZ and CBP primers suggested that the inv(8) does not result in a cryptic MOZ-CBP fusion. It is likely, therefore, that MOZ is fused to a novel gene at 8q13 in this case. We conclude that the t(8;13) breakpoint is flanked by YACs 782H11 and 847B12 and is at least 1 Mb telomeric to MOZ. MOZ is involved, however, in a new variant of the t(8;16).


Subject(s)
Acetyltransferases/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 16 , Chromosomes, Human, Pair 8 , Leukemia, Monocytic, Acute/genetics , Leukemia, Myelomonocytic, Acute/genetics , Lymphoma, Non-Hodgkin/genetics , Nuclear Proteins/genetics , Trans-Activators , Transcription Factors/genetics , Blotting, Southern , CREB-Binding Protein , Chromosome Inversion , Chromosome Mapping , Histone Acetyltransferases , Humans , In Situ Hybridization, Fluorescence , Polymerase Chain Reaction , Translocation, Genetic
20.
Leukemia ; 11(2): 233-8, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9009086

ABSTRACT

Chromosome band 9p21-22 is one of the most common targets for deletions in cancer. The p16 tumor suppressor gene maps to this region and is inactivated in a wide variety of tumor cell lines and primary tumors. However, in some of the neoplasms with frequent 9p21 loss of heterozygosity (LOH), a structurally and functionally normal p16 is found suggesting that this gene might not be the primary or only target for inactivation. To define the smallest region of overlap of deletions at chromosome band 9p21-22 and to clarify the involvement of p16, p15 and the IFN cluster gene in lymphoblastic leukemias, we used a multiplex polymerase chain reaction to construct a detailed map of deletions at 9p21-22. We studied DNA from 30 lymphoblastic leukemia patients and nine cell lines using 10 genes/markers that map to this region, including four STS markers located between p16 and D9S171 (STS1, STS2) or between p16 and IFNalpha (STS3, STS4). We found that the size of the deletions in this region is variable and that the commonly deleted region spans at least 400 kb; it includes the p16 gene but excludes the IFN gene cluster and the p15 gene. The identification of this commonly deleted region enables a more focused search for other putative tumor suppressor gene at 9p21 which could be relevant to leukemogenesis.


Subject(s)
Carrier Proteins/genetics , Cell Cycle Proteins , Chromosomes, Human, Pair 9/genetics , Genes, Tumor Suppressor , Leukemia, Myeloid/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Sequence Deletion , Tumor Suppressor Proteins , Acute Disease , Chromosomes, Human, Pair 9/ultrastructure , Chronic Disease , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16 , DNA, Neoplasm/genetics , Heterozygote , Humans , Interferon-alpha/genetics , Polymerase Chain Reaction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL