Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
2.
Leukemia ; 29(12): 2317-27, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26108691

ABSTRACT

The TLX1 transcription factor is critically involved in the multi-step pathogenesis of T-cell acute lymphoblastic leukemia (T-ALL) and often cooperates with NOTCH1 activation during malignant T-cell transformation. However, the exact molecular mechanism by which these T-cell specific oncogenes cooperate during transformation remains to be established. Here, we used chromatin immunoprecipitation followed by sequencing to establish the genome-wide binding pattern of TLX1 in human T-ALL. This integrative genomics approach showed that ectopic TLX1 expression drives repression of T cell-specific enhancers and mediates an unexpected transcriptional antagonism with NOTCH1 at critical target genes, including IL7R and NOTCH3. These phenomena coordinately trigger a TLX1-driven pre-leukemic phenotype in human thymic precursor cells, reminiscent of the thymus regression observed in murine TLX1 tumor models, and create a strong genetic pressure for acquiring activating NOTCH1 mutations as a prerequisite for full leukemic transformation. In conclusion, our results uncover a functional antagonism between cooperative oncogenes during the earliest phases of tumor development and provide novel insights in the multi-step pathogenesis of TLX1-driven human leukemia.


Subject(s)
Homeodomain Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Homeodomain Proteins/physiology , Humans , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Proto-Oncogene Proteins/physiology , Receptor, Notch1/genetics , Receptor, Notch1/physiology
3.
Leukemia ; 29(4): 798-806, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25231743

ABSTRACT

The MYB oncogene is a leucine zipper transcription factor essential for normal and malignant hematopoiesis. In T-cell acute lymphoblastic leukemia (T-ALL), elevated MYB levels can arise directly through T-cell receptor-mediated MYB translocations, genomic MYB duplications or enhanced TAL1 complex binding at the MYB locus or indirectly through the TAL1/miR-223/FBXW7 regulatory axis. In this study, we used an unbiased MYB 3'untranslated region-microRNA (miRNA) library screen and identified 33 putative MYB-targeting miRNAs. Subsequently, transcriptome data from two independent T-ALL cohorts and different subsets of normal T-cells were used to select miRNAs with relevance in the context of normal and malignant T-cell transformation. Hereby, miR-193b-3p was identified as a novel bona fide tumor-suppressor miRNA that targets MYB during malignant T-cell transformation thereby offering an entry point for efficient MYB targeting-oriented therapies for human T-ALL.


Subject(s)
Gene Expression Regulation, Leukemic , MicroRNAs/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-myb/genetics , T-Lymphocytes/metabolism , 3' Untranslated Regions , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , F-Box Proteins/genetics , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Gene Expression Profiling , Genomic Library , Humans , Mice , MicroRNAs/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Primary Cell Culture , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Signal Transduction , T-Cell Acute Lymphocytic Leukemia Protein 1 , T-Lymphocytes/pathology , Transcriptome , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
4.
Br J Cancer ; 112(2): 290-5, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25356804

ABSTRACT

BACKGROUND: The prognostic impact of segmental chromosome alterations (SCAs) in children older than 1 year, diagnosed with localised unresectable neuroblastoma (NB) without MYCN amplification enrolled in the European Unresectable Neuroblastoma (EUNB) protocol is still to be clarified, while, for other group of patients, the presence of SCAs is associated with poor prognosis. METHODS: To understand the role of SCAs we performed multilocus/pangenomic analysis of 98 tumour samples from patients enrolled in the EUNB protocol. RESULTS: Age at diagnosis was categorised into two groups using 18 months as the age cutoff. Significant difference in the presence of SCAs was seen in tumours of patients between 12 and 18 months and over 18 months of age at diagnosis, respectively (P=0.04). A significant correlation (P=0.03) was observed between number of SCAs per tumour and age. Event-free (EFS) and overall survival (OS) were calculated in both age groups, according to both the presence and number of SCAs. In older patients, a poorer survival was associated with the presence of SCAs (EFS=46% vs 75%, P=0.023; OS=66.8% vs 100%, P=0.003). Moreover, OS of older patients inversely correlated with number of SCAs (P=0.002). Finally, SCAs provided additional prognostic information beyond histoprognosis, as their presence was associated with poorer OS in patients over 18 months with unfavourable International Neuroblastoma Pathology Classification (INPC) histopathology (P=0.018). CONCLUSIONS: The presence of SCAs is a negative prognostic marker that impairs outcome of patients over the age of 18 months with localised unresectable NB without MYCN amplification, especially when more than one SCA is present. Moreover, in older patients with unfavourable INPC tumour histoprognosis, the presence of SCAs significantly affects OS.


Subject(s)
Neuroblastoma/genetics , Peripheral Nervous System Neoplasms/genetics , Chromosome Aberrations , Comparative Genomic Hybridization , Disease-Free Survival , Gene Amplification , Humans , Infant , Kaplan-Meier Estimate , N-Myc Proto-Oncogene Protein , Neuroblastoma/diagnosis , Neuroblastoma/mortality , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Peripheral Nervous System Neoplasms/diagnosis , Peripheral Nervous System Neoplasms/mortality , Prognosis
5.
Int J Cancer ; 130(11): 2599-606, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-21796619

ABSTRACT

Neuroblastoma is an aggressive embryonal tumor that accounts for ∼15% of childhood cancer deaths. Hitherto, despite the availability of comprehensive genomic data on DNA copy number changes in neuroblastoma, relatively little is known about the genes driving neuroblastoma tumorigenesis. In this study, high resolution array comparative genome hybridization (CGH) was performed on 188 primary neuroblastoma tumors and 33 neuroblastoma cell lines to search for previously undetected recurrent DNA copy number gains and losses. A new recurrent distal chromosome 1q deletion (del(1)(q42.2qter)) was detected in seven cases. Further analysis of available array CGH datasets revealed 13 additional similar distal 1q deletions. The majority of all detected 1q deletions was found in high risk 11q deleted tumors without MYCN amplification (Fisher exact test p = 5.61 × 10(-5) ). Using ultra-high resolution (∼115 bp resolution) custom arrays covering the breakpoints on 1q for 11 samples, clustering of nine breakpoints was observed within a 12.5-kb region, of which eight were found in a 7-kb copy number variable region, whereas the remaining two breakpoints were colocated 1.4-Mb proximal. The commonly deleted region contains one miRNA (hsa-mir-1537), four transcribed ultra conserved region elements (uc.43-uc.46) and 130 protein coding genes including at least two bona fide tumor suppressor genes, EGLN1 (or PHD2) and FH. This finding further contributes to the delineation of the genomic profile of aggressive neuroblastoma, offers perspectives for the identification of genes contributing to the disease phenotype and may be relevant in the light of assessment of response to new molecular treatments.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 11 , Gene Dosage , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Cell Line, Tumor , Comparative Genomic Hybridization , Fumarate Hydratase/genetics , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , N-Myc Proto-Oncogene Protein , Procollagen-Proline Dioxygenase/genetics
6.
Br J Cancer ; 105(12): 1940-8, 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-22146831

ABSTRACT

BACKGROUND: In neuroblastoma (NB), the presence of segmental chromosome alterations (SCAs) is associated with a higher risk of relapse. METHODS: In order to analyse the role of SCAs in infants with localised unresectable/disseminated NB without MYCN amplification, we have performed an array CGH analysis of tumours from infants enrolled in the prospective European INES trials. RESULTS: Tumour samples from 218 out of 300 enroled patients could be analysed. Segmental chromosome alterations were observed in 11%, 20% and 59% of infants enroled in trials INES99.1 (localised unresectable NB), INES99.2 (stage 4s) and INES99.3 (stage 4) (P<0.0001). Progression-free survival was poorer in patients whose tumours harboured SCA, in the whole population and in trials INES99.1 and INES99.2, in the absence of clinical symptoms (log-rank test, P=0.0001, P=0.04 and P=0.0003, respectively). In multivariate analysis, a SCA genomic profile was the strongest predictor of poorer progression-free survival. CONCLUSION: In infants with stage 4s MYCN-non-amplified NB, a SCA genomic profile identifies patients who will require upfront treatment even in the absence of other clinical indication for therapy, whereas in infants with localised unresectable NB, a genomic profile characterised by the absence of SCA identifies patients in whom treatment reduction might be possible. These findings will be implemented in a future international trial.


Subject(s)
Chromosome Aberrations , Neuroblastoma/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Humans , Infant , N-Myc Proto-Oncogene Protein , Neuroblastoma/genetics , Prognosis , Prospective Studies , Recurrence , Survival Analysis
7.
Oncogene ; 29(9): 1394-404, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-19946337

ABSTRACT

Increased activity of MYC protein-family members is a common feature in many cancers. Using neuroblastoma as a tumor model, we established a microRNA (miRNA) signature for activated MYCN/c-MYC signaling in two independent primary neuroblastoma tumor cohorts and provide evidence that c-MYC and MYCN have overlapping functions. On the basis of an integrated approach including miRNA and messenger RNA (mRNA) gene expression data we show that miRNA activation contributes to widespread mRNA repression, both in c-MYC- and MYCN-activated tumors. c-MYC/MYCN-induced miRNA activation was shown to be dependent on c-MYC/MYCN promoter binding as evidenced by chromatin immunoprecipitation. Finally, we show that pathways, repressed through c-MYC/MYCN miRNA activation, are highly correlated to tumor aggressiveness and are conserved across different tumor entities suggesting that c-MYC/MYCN activate a core set of miRNAs for cooperative repression of common transcriptional programs related to disease aggressiveness. Our results uncover a widespread correlation between miRNA activation and c-MYC/MYCN-mediated coding gene expression modulation and further substantiate the overlapping functions of c-MYC and MYCN in the process of tumorigenesis.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Gene Regulatory Networks/drug effects , Genes, myc/physiology , MicroRNAs/pharmacology , Neuroblastoma/genetics , Nuclear Proteins/pharmacology , Oncogene Proteins/pharmacology , Promoter Regions, Genetic/drug effects , Cell Line, Tumor , Gene Regulatory Networks/physiology , Gene Silencing/physiology , Genes, myc/genetics , Humans , MicroRNAs/biosynthesis , N-Myc Proto-Oncogene Protein , Neuroblastoma/therapy , RNA, Small Interfering/pharmacology , Transcription Factors/physiology , Treatment Outcome
8.
Verh K Acad Geneeskd Belg ; 69(4): 167-96, 2007.
Article in Dutch | MEDLINE | ID: mdl-17821957

ABSTRACT

Neuroblastoma (NB) is, next to acute lymphoblastic leukaemia, brain tumours and lymphoma the most frequent paediatric tumour (8-10%). Our research group aims to contribute to the unravelling of the genetic basis of NB. Insight into the genes and signalling pathways involved in tumour formation and development can represent an essential step towards the development of more efficient molecular targeted therapies. A first part of our research work was devoted to the analysis of genomic alterations in NB. By means of a new highly sensitive method for detecting gains and losses of chromosomal segments, we recognised three major prognostic relevant genomic subtypes of NB. In addition smaller subgroups with deviating genomic patterns were detected. In addition, this work yielded important information regarding delineation of critical regions of gain and loss in NB which should facilitate further selection of candidate oncogenes or tumour suppressor genes. A second important part of our work focussed on the gene expression profiling of NB precursor cells. We were able as the first to isolate these cells and determine their transcriptome, a finding of fundamental importance for future expression studies in NB. Another study focussed on the identification of MYCN transcriptional target genes. Gene expression analyses of model systems developed in our lab and of a large panel of cell lines and tumours allowed us to subtract a list of candidate genes which are now under further study. Finally, we initiated research towards the understanding of the role of methylation in NB oncogenesis. From this, we were able to create a list of potentially relevant methylated genes in NB. From the above it is clear that our team has made important contributions to the understanding of the complex biology and clinical behaviour of NB. Also, a broad technically innovative research platform has been developed which will allow us to dissect NB genetics with greater speed and accuracy.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Neuroblastoma/genetics , Genes, Neoplasm , Humans , Neuroblastoma/pathology
9.
Leukemia ; 21(1): 121-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17039236

ABSTRACT

Recently, we and others described a new chromosomal rearrangement, that is, inv(7)(p15q34) and t(7;7)(p15;q34) involving the T-cell receptor beta (TCRbeta) (7q34) and the HOXA gene locus (7p15) in 5% of T-cell acute lymphoblastic leukemia (T-ALL) patients leading to transcriptional activation of especially HOXA10. To further address the clinical, immunophenotypical and molecular genetic findings of this chromosomal aberration, we studied 330 additional T-ALLs. This revealed TCRbeta-HOXA rearrangements in five additional patients, which brings the total to 14 cases in 424 patients (3.3%). Real-time quantitative PCR analysis for HOXA10 gene expression was performed in 170 T-ALL patients and detected HOXA10 overexpression in 25.2% of cases including all the cases with a TCRbeta-HOXA rearrangement (8.2%). In contrast, expression of the short HOXA10 transcript, HOXA10b, was almost exclusively found in the TCRbeta-HOXA rearranged cases, suggesting a specific role for the HOXA10b short transcript in TCRbeta-HOXA-mediated oncogenesis. Other molecular and/or cytogenetic aberrations frequently found in subtypes of T-ALL (SIL-TAL1, CALM-AF10, HOX11, HOX11L2) were not detected in the TCRbeta-HOXA rearranged cases except for deletion 9p21 and NOTCH1 activating mutations, which were present in 64 and 67%, respectively. In conclusion, this study defines TCRbeta-HOXA rearranged T-ALLs as a distinct cytogenetic subgroup by clinical, immunophenotypical and molecular genetic characteristics.


Subject(s)
Homeodomain Proteins/genetics , Leukemia-Lymphoma, Adult T-Cell/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Adolescent , Adult , Child , Chromosome Deletion , Chromosome Inversion , Female , Gene Rearrangement, T-Lymphocyte , Homeobox A10 Proteins , Humans , Immunophenotyping , Leukemia-Lymphoma, Adult T-Cell/pathology , Leukemia-Lymphoma, Adult T-Cell/physiopathology , Male , Middle Aged , Receptor, Notch1/genetics , Transcriptional Activation , Translocation, Genetic
10.
Cytogenet Genome Res ; 115(3-4): 273-82, 2006.
Article in English | MEDLINE | ID: mdl-17124410

ABSTRACT

In the past few years high throughput methods for assessment of DNA copy number alterations have witnessed rapid progress. Both 'in house' developed BAC, cDNA, oligonucleotide and commercial arrays are now available and widely applied in the study of the human genome, particularly in the context of disease. Cancer cells are known to exhibit DNA losses, gains and amplifications affecting tumor suppressor genes and proto-oncogenes. Moreover, these patterns of genomic imbalances may be associated with particular tumor types or subtypes and may have prognostic value. Here we summarize recent array CGH findings in neuroblastoma, a pediatric tumor of the sympathetic nervous system. A total of 176 primary tumors and 53 cell lines have been analyzed on different platforms. Through these studies the genomic content and boundaries of deletions, gains and amplifications were characterized with unprecedented accuracy. Furthermore, in conjunction with cytogenetic findings, array CGH allows the mapping of breakpoints of unbalanced translocations at a very high resolution.


Subject(s)
Computational Biology/methods , DNA/analysis , Genome, Human , Neuroblastoma/genetics , Cell Line, Tumor , DNA, Neoplasm , Humans , In Situ Hybridization, Fluorescence , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Oligonucleotides/chemistry , Oligonucleotides/genetics , Translocation, Genetic
12.
Leukemia ; 19(3): 358-66, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15674412

ABSTRACT

Chromosomal translocations with breakpoints in T-cell receptor (TCR) genes are recurrent in T-cell malignancies. These translocations involve the TCRalphadelta gene (14q11), the TCRbeta gene (7q34) and to a lesser extent the TCRgamma gene at chromosomal band 7p14 and juxtapose T-cell oncogenes next to TCR regulatory sequences leading to deregulated expression of those oncogenes. Here, we describe a new recurrent chromosomal inversion of chromosome 7, inv(7)(p15q34), in a subset of patients with T-cell acute lymphoblastic leukemia characterized by CD2 negative and CD4 positive, CD8 negative blasts. This rearrangement juxtaposes the distal part of the HOXA gene cluster on 7p15 to the TCRbeta locus on 7q34. Real time quantitative PCR analysis for all HOXA genes revealed high levels of HOXA10 and HOXA11 expression in all inv(7) positive cases. This is the first report of a recurrent chromosome rearrangement targeting the HOXA gene cluster in T-cell malignancies resulting in deregulated HOXA gene expression (particularly HOXA10 and HOXA11) and is in keeping with a previous report suggesting HOXA deregulation in MLL-rearranged T- and B cell lymphoblastic leukemia as the key factor in leukaemic transformation. Finally, our observation also supports the previous suggested role of HOXA10 and HOXA11 in normal thymocyte development.


Subject(s)
Chromosome Inversion , Chromosomes, Human, Pair 7/genetics , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Leukemia-Lymphoma, Adult T-Cell/genetics , Transcriptional Activation/genetics , Adolescent , Adult , Child , Child, Preschool , Cytogenetic Analysis , DNA-Binding Proteins/physiology , Female , Gene Expression Regulation, Neoplastic , Gene Rearrangement, T-Lymphocyte/genetics , Homeobox A10 Proteins , Homeodomain Proteins/physiology , Humans , Immunophenotyping , Male , Middle Aged , Translocation, Genetic/genetics
13.
Eur J Cancer ; 40(8): 1255-61, 2004 May.
Article in English | MEDLINE | ID: mdl-15110891

ABSTRACT

Although neuroblastoma is the most common extracranial solid tumour of childhood, little is known about its aetiology. Together with MYCN amplification and chromosome 17q gain, chromosome 1p deletion is one of the most frequently occurring genetic abnormalities in neuroblastoma. Based upon mapping of deletion breakpoints, putative tumour suppressor gene loci have been assigned to the distal part of the short arm of chromosome 1. Recently, the EXTL1 gene was suggested as a candidate neuroblastoma-suppressor gene and to evaluate this hypothesis, we performed 1p deletion analysis and mutation screening of the EXTL1-coding region on DNA from 22 primary neuroblastomas and 21 neuroblastoma cell lines. Deletions of the chromosome region 1p36.1, including the EXTL1 gene, were detected in several neuroblastoma cell lines and primary tumours. EXTL1 mutation screening resulted in the detection of one unclassified variant (Ser28Cys) but could not provide additional evidence of EXTL1 being involved in the aetiology of neuroblastoma.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 1/genetics , Mutation/genetics , N-Acetylglucosaminyltransferases/genetics , Neuroblastoma/genetics , Tumor Suppressor Proteins/genetics , Cell Line, Tumor , Humans , Polymorphism, Genetic
14.
J Clin Oncol ; 21(11): 2077-84, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12775732

ABSTRACT

PURPOSE: Therapy stratification based on genetic markers is becoming increasingly important, which makes commitment to the highest possible reliability of the involved markers mandatory. In neuroblastic tumors, amplification of the MYCN gene is an unequivocal marker that indicates aggressive tumor behavior and is consequently used for therapy stratification. To guarantee reliable and standardized quality of genetic features, a quality-assessment study was initiated by the European Neuroblastoma Quality Assessment (ENQUA; connected to International Society of Pediatric Oncology) Group. MATERIALS AND METHODS: One hundred thirty-seven coded specimens from 17 tumors were analyzed in 11 European national/regional reference laboratories using molecular techniques, in situ hybridization, and flow and image cytometry. Tumor samples with divergent results were re-evaluated. RESULTS: Three hundred fifty-two investigations were performed, which resulted in 23 divergent findings, 17 of which were judged as errors after re-evaluation. MYCN analyses determined by Southern blot and in situ hybridization led to 3.7% and 4% of errors, respectively. Tumor cell content was not indicated in 32% of the samples, and 11% of seemingly correct MYCN results were based on the investigation of normal cells (eg, Schwann cells). Thirty-eight investigations were considered nonassessable. CONCLUSION: This study demonstrated the importance of revealing the difficulties and limitations for each technique and problems in interpreting results, which are crucial for therapeutic decisions. Moreover, it led to the formulation of guidelines that are applicable to all kinds of tumors and that contain the standardization of techniques, including the exact determination of the tumor cell content. Finally, the group has developed a common terminology for molecular-genetic results.


Subject(s)
Biomarkers, Tumor/analysis , Genetic Techniques/standards , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Quality Assurance, Health Care , Biomarkers, Tumor/genetics , Blotting, Southern , Chromosomes, Human, Pair 1/genetics , DNA, Neoplasm/analysis , Diagnostic Errors/prevention & control , Diagnostic Errors/statistics & numerical data , Europe , Humans , In Situ Hybridization, Fluorescence , N-Myc Proto-Oncogene Protein , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Ploidies , Polymerase Chain Reaction , Quality Control , Reference Standards , Terminology as Topic
15.
Genes Chromosomes Cancer ; 32(2): 126-35, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11550280

ABSTRACT

Cancer cell lines are essential gene discovery tools and have often served as models in genetic and functional studies of particular tumor types. One of the future challenges is comparison and interpretation of gene expression data with the available knowledge on the genomic abnormalities in these cell lines. In this context, accurate description of these genomic abnormalities is required. Here, we show that a combination of M-FISH with banding analysis, standard FISH, and CGH allowed a detailed description of the genetic alterations in 16 neuroblastoma cell lines. In total, 14 cryptic chromosome rearrangements were detected, including a balanced t(2;4)(p24.3;q34.3) translocation in cell line NBL-S, with the 2p24 breakpoint located at about 40 kb from MYCN. The chromosomal origin of 22 marker chromosomes and 41 cytogenetically undefined translocated segments was determined. Chromosome arm 2 short arm translocations were observed in six cell lines (38%) with and five (31%) without MYCN amplification, leading to partial chromosome arm 2p gain in all but one cell line and loss of material in the various partner chromosomes, including 1p and 11q. These 2p gains were often masked in the GGH profiles due to MYCN amplification. The commonly overrepresented region was chromosome segment 2pter-2p22, which contains the MYCN gene, and five out of eleven 2p breakpoints clustered to the interface of chromosome bands 2p16 and 2p21. In neuroblastoma cell line SJNB-12, with double minutes (dmins) but no MYCN amplification, the dmins were shown to be derived from 16q22-q23 sequences. The ATBF1 gene, an AT-binding transcription factor involved in normal neurogenesis and located at 16q22.2, was shown to be present in the amplicon. This is the first report describing the possible implication of ATBF1 in neuroblastoma cells. We conclude that a combined approach of M-FISH, cytogenetics, and CGH allowed a more complete and accurate description of the genetic alterations occurring in the investigated cell lines.


Subject(s)
Chromosome Painting/methods , In Situ Hybridization, Fluorescence/methods , Neuroblastoma/genetics , Nucleic Acid Hybridization , Female , Humans , Karyotyping , Male , Nucleic Acid Hybridization/methods , Tumor Cells, Cultured
16.
Cancer Genet Cytogenet ; 128(1): 39-42, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11454428

ABSTRACT

Bloom syndrome (BS) predisposes affected individuals to a wide variety of neoplasms including hematological malignancies. Thus far, cytogenetic findings in hematological neoplasms have been reported in only a few BS patients. We present the karyotypic findings in a BS patient diagnosed with acute myeloid leukemia (AML), FAB subtype M1, and a review of the literature, showing the preferential occurrence of total or partial loss of chromosome 7 in BS patients with AML or myelodysplastic syndromes (MDS).


Subject(s)
Bloom Syndrome/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 7/genetics , Leukemia, Myeloid/genetics , Myelodysplastic Syndromes/genetics , Acute Disease , Adolescent , Adult , Child , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male
17.
Med Pediatr Oncol ; 36(1): 20-3, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11464884

ABSTRACT

BACKGROUND: Unbalanced translocations resulting in the gain of material from 17q are the most common chromosomal changes in neuroblastoma and are associated with poor patient survival, and are established indicators of bad prognosis. PROCEDURE: We have used 13 fluorescent in situ hybridisation probes to map 17q translocation breakpoints in ten neuroblastoma cell lines and 21 primary tumours. RESULTS: At least seven different breakpoints have been identified, all localised within the proximal half of 17q (53-68 cM, 17cen-17q22). CONCLUSION: These results suggest that the dosage of a gene, or genes, in 17q22-qter is responsible for the clinical effects of 17q gain, rather than the disruption of a specific gene.


Subject(s)
Chromosome Breakage , Chromosomes, Human, Pair 17/ultrastructure , Neuroblastoma/genetics , Translocation, Genetic , Chromosome Mapping , Chromosomes, Human, Pair 17/genetics , DNA Probes , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Neuroblastoma/mortality , Neuroblastoma/ultrastructure , Prognosis , Tumor Cells, Cultured/ultrastructure
18.
Med Pediatr Oncol ; 36(1): 5-10, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11464905

ABSTRACT

PROCEDURE: Analysis of comparative genomic hybridization (CGH) data of 120 tumors from four different studies, and data of 84 previously unpublishied tumors, allowed delineation of at least six different genetic subsets of neuroblastomas. RESULTS AND CONCLUSIONS: A small number of tumors show no detectable imballances. A second group of tumors presents with gains and losses of whole chromosomes and is found predominantly in prognostically favorable stage 1 and 2 tumors. The remaining groups are characterized by the presence of partial chromosome imbalances, and are found mostly in stage 3, 4, and 4S tumors. The third group shows 17q gain without 11q loss, 1p loss, or MYCN amplification (MNA). The fourth group has 1p deletion or MNA, and finally, a fifth group shows 11q loss without 1p deletion or MNA, and is found mainly in stage 4 tumors. The latter group is significantly associated with losses of 3p, 4p, and 14q.


Subject(s)
Chromosome Aberrations , DNA, Neoplasm/analysis , Neuroblastoma/genetics , Chromosome Deletion , Chromosomes, Human/genetics , Chromosomes, Human/ultrastructure , Humans , Loss of Heterozygosity , Neoplasm Staging , Neuroblastoma/classification , Neuroblastoma/mortality , Nucleic Acid Hybridization , Prognosis , Trisomy
19.
Int J Cancer ; 92(3): 409-13, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11291079

ABSTRACT

Merkel cell carcinoma (MCC) is a rare, highly metastatic skin tumor of neuroectodermal origin. The disease shares clinical and histopathological features with small cell lung carcinoma (SCLC). The genetic mechanisms underlying the development and tumor progression of MCC are poorly understood. We recently showed by comparative genomic hybridization (CGH) that the pattern of chromosomal abnormalities in MCC resembles that of SCLC. One of the most frequently observed losses involved the entire chromosome 10 or partial loss of the chromosome 10 long arm (33% of examined MCC cases). The PTEN tumor-suppressor gene has been mapped to 10q23.3 and was shown to be mutated in a variety of human cancers including SCLC. Germline PTEN mutations have been observed in familial predisposing cancer syndromes including Cowden disease. Interestingly, an association between Cowden syndrome and Merkel cell carcinoma has been reported. To study the possible role of PTEN in MCC oncogenesis, loss of heterozygosity (LOH) analysis for the 10q23 region was performed on 26 MCC tumor samples from 23 MCC patients. The PTEN locus was deleted in 9 of 21 (43%) informative MCC tumor samples [7 of 18 (39%) MCC patients]. Despite this high frequency of LOH at 10q23, mutation and homozygous deletion screening of the PTEN gene revealed only one tumor with a nonsense mutation and a second with a homozygous deletion of exon 9. These data suggest that either alternative mechanisms lead to inactivation of the PTEN gene or that other tumor-suppressor genes at chromosome 10 are implicated in the development of MCC.


Subject(s)
Carcinoma, Merkel Cell/genetics , Chromosomes, Human, Pair 10 , Loss of Heterozygosity/genetics , Phosphoric Monoester Hydrolases/genetics , Skin Neoplasms/genetics , Tumor Suppressor Proteins , Aged , Aged, 80 and over , DNA Mutational Analysis , Female , Gene Deletion , Genetic Testing , Humans , Male , Middle Aged , Mutation , PTEN Phosphohydrolase
20.
Int J Cancer ; 91(5): 680-6, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11267980

ABSTRACT

We have studied the occurrence and association of 11q deletions with other chromosomal imbalances in Stage 4 neuroblastomas. To this purpose we have performed comparative genomic hybridization (CGH) analysis on 50 Stage 4 neuroblastomas and these data were analyzed together with those from 33 previously published cases. We observed a high incidence of 11q deletion in Stage 4 neuroblastoma without MYCN amplification (59%) whereas 11q loss was only observed in 15% of neuroblastomas with MYCN-amplification (p = 0.0002) or 11% of cases with 1p deletion detected by CGH (p = 0.0001). In addition, 11q loss showed significant positive correlation with 3p loss (p = 0.0002). Event-free survival was poor and not significantly different for patients with or without 11q deletion. Our study provides further evidence that Stage 4 neuroblastomas with 11q deletions represent a distinct genetic subgroup that typically shows no MYCN-amplification nor 1p deletion. Moreover, it shows that neuroblastomas with 11q deletion also often present 3p deletion. This genetic subgroup shows a similar poor prognosis as MYCN amplified 4 neuroblastomas.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 11 , Genome, Human , Neuroblastoma/genetics , Nucleic Acid Hybridization , Adolescent , Child , Child, Preschool , Chromosome Aberrations , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 3 , Disease-Free Survival , Female , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Models, Genetic , Multicenter Studies as Topic , Mutation , Neoplasm Metastasis , Neuroblastoma/diagnosis , Neuroblastoma/mortality , Prognosis , Time Factors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...