Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
2.
J Mol Diagn ; 15(1): 51-61, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23246132

ABSTRACT

Anaplastic lymphoma kinase gene (ALK) fusions have been identified in approximately 5% of non-small-cell lung carcinomas (NSCLCs) and define a distinct subpopulation of patients with lung cancer who are highly responsive to ALK kinase inhibitors, such as crizotinib. Because of this profound therapeutic implication, the latest National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology recommend upfront ALK screening for all patients with NSCLC. The Food and Drug Administration-approved companion diagnostic test (ie, fluorescence in situ hybridization) for identification of ALK-positive patients, however, is complex and has considerable limitations in terms of cost and throughput, making it difficult to screen many patients. To explore alternative screening modalities for detecting ALK fusions, we designed a combination of two transcript-based assays to detect for presence or absence of ALK fusions using NanoString's nCounter technology. By using this combined gene expression and ALK fusion detection strategy, we developed a multiplexed assay with a quantitative scoring modality that is highly sensitive, reproducible, and capable of detecting low-abundant ALK fusion transcripts, even in samples with a low tumor cell content. In 66 archival NSCLC samples, our results were highly concordant to prior results obtained by fluorescence in situ hybridization and IHC. Our assay offers a cost-effective, easy-to-perform, high-throughput, and FFPE-compatible screening alternative for detection of ALK fusions.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression , Lung Neoplasms/genetics , Oncogene Fusion , Receptor Protein-Tyrosine Kinases/genetics , Anaplastic Lymphoma Kinase , Base Sequence , Carcinoma, Non-Small-Cell Lung/diagnosis , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Lung Neoplasms/diagnosis , Molecular Sequence Data , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/isolation & purification , Polymerase Chain Reaction , Reproducibility of Results , Sequence Analysis, DNA
3.
Genes Chromosomes Cancer ; 52(1): 11-23, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22965931

ABSTRACT

Uterine leiomyomas are benign solid tumors of mesenchymal origin which occur with an estimated incidence of up to 77% of all women of reproductive age. The majority of these tumors remains symptomless, but in about a quarter of cases they cause leiomyoma-associated symptoms including chronic pelvic pain, menorrhagia-induced anemia, and impaired fertility. As a consequence, they are the most common indication for pre-menopausal hysterectomy in the USA and Japan and annually translate into a multibillion dollar healthcare problem. Approximately 40% of these neoplasms present with recurring structural cytogenetic anomalies, including del(7)(q22), t(12;14)(q15;q24), t(1;2)(p36;p24), and anomalies affecting 6p21 and/or 10q22. Using positional cloning strategies, we and others previously identified HMGA1, HMGA2, RAD51L1, MORF, and, more recently, NCOA1 as primary target (fusion) genes associated with tumor initiation in four of these distinct cytogenetic subgroups. Despite the fact that the del(7)(q22) subgroup is the largest among leiomyomas, and was first described more than twenty years ago, the 7q22 leiomyoma target gene still awaits unequivocal identification. We here describe a positional cloning effort from two independent uterine leiomyomas, containing respectively a pericentric and a paracentric chromosomal inversion, both affecting band 7q22. We found that both chromosomal inversions target the cut-like homeobox 1 (CUX1) gene on chromosomal band 7q22.1 in a way which is functionally equivalent to the more frequently observed del(7q) cases, and which is compatible with a mono-allelic knock-out scenario, similar as was previously described for the cytogenetic subgroup showing chromosome 14q involvement.


Subject(s)
Biomarkers, Tumor/genetics , Chromosomes, Human, Pair 7 , Homeodomain Proteins/genetics , Leiomyoma/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Uterine Neoplasms/genetics , Amino Acid Sequence , Base Sequence , Female , Humans , In Situ Hybridization, Fluorescence , Middle Aged , Molecular Sequence Data , Transcription Factors
4.
Hum Genet ; 128(3): 281-91, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20563892

ABSTRACT

Microcephaly, mental retardation and congenital retinal folds along with other systemic features have previously been reported as a separate clinical entity. The sporadic nature of the syndrome and lack of clear inheritance patterns pointed to a genetic heterogeneity. Here, we report a genetic analysis of a female patient with microcephaly, congenital bilateral falciform retinal folds, nystagmus, and mental retardation. Karyotyping revealed a de novo pericentric inversion in chromosome 6 with breakpoints in 6p12.1 and 6q21. Fluorescence in situ hybridization analysis narrowed down the region around the breakpoints, and the breakpoint at 6q21 was found to disrupt the CDK19 gene. CDK19 was found to be expressed in a diverse range of tissues including fetal eye and fetal brain. Quantitative PCR of the CDK19 transcript from Epstein-Barr virus-transformed lymphoblastoid cell lines of the patient revealed ~50% reduction in the transcript (p = 0.02), suggesting haploinsufficiency of the gene. cdk8, the closest orthologue of human CDK19 in Drosophila has been shown to play a major role in eye development. Conditional knock-down of Drosophila cdk8 in multiple dendrite (md) neurons resulted in 35% reduced dendritic branching and altered morphology of the dendritic arbour, which appeared to be due in part to a loss of small higher order branches. In addition, Cdk8 mutant md neurons showed diminished dendritic fields revealing an important role of the CDK19 orthologue in the developing nervous system of Drosophila. This is the first time the CDK19 gene, a component of the mediator co-activator complex, has been linked to a human disease.


Subject(s)
Abnormalities, Multiple/genetics , Cyclin-Dependent Kinases/genetics , Intellectual Disability/genetics , Microcephaly/genetics , Retina/abnormalities , Adult , Animals , Base Sequence , Chromosome Inversion , Chromosomes, Human, Pair 6/genetics , Cyclin-Dependent Kinase 8/genetics , DNA Primers/genetics , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Species Specificity
5.
Am J Hum Genet ; 86(1): 72-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20004881

ABSTRACT

Next-generation sequencing is a straightforward tool for the identification of disease genes in extended genomic regions. Autozygosity mapping was performed on a five-generation inbred Italian family with three siblings affected with Clericuzio-type poikiloderma with neutropenia (PN [MIM %604173]), a rare autosomal-recessive genodermatosis characterised by poikiloderma, pachyonychia, and chronic neutropenia. The siblings were initially diagnosed as affected with Rothmund-Thomson syndrome (RTS [MIM #268400]), with which PN shows phenotypic overlap. Linkage analysis on all living subjects of the family identified a large 16q region inherited identically by descent (IBD) in all affected family members. Deep sequencing of this 3.4 Mb region previously enriched with array capture revealed a homozygous c.504-2 A>C mismatch in all affected siblings. The mutation destroys the invariant AG acceptor site of intron 4 of the evolutionarily conserved C16orf57 gene. Two distinct deleterious mutations (c.502A>G and c.666_676+1del12) identified in an unrelated PN patient confirmed that the C16orf57 gene is responsible for PN. The function of the predicted C16orf57 gene is unknown, but its product has been shown to be interconnected to RECQL4 protein via SMAD4 proteins. The unravelled clinical and genetic identity of PN allows patients to undergo genetic testing and follow-up.


Subject(s)
Neutropenia/genetics , Open Reading Frames , Sequence Analysis, DNA/methods , Skin Diseases/genetics , Abnormalities, Multiple/genetics , DNA Mutational Analysis , Diagnosis, Differential , Evolution, Molecular , Female , Genomics , Genotype , Heterozygote , Homozygote , Humans , Male , Pedigree , Rothmund-Thomson Syndrome/genetics
6.
Cancer Genet Cytogenet ; 195(2): 105-11, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19963109

ABSTRACT

FBXW7 (alias CDC4) is a p53-dependent tumor suppressor gene that exhibits mutations or deletions in a variety of human tumors. Mutation or deletion of the FBXW7 gene has been associated with an increase in chromosomal instability and cell cycle progression. In addition, the FBXW7 protein has been found to act as a component of the ubiquitin proteasome system and to degrade several oncogenic proteins that function in cellular growth regulatory pathways. By using a rapid breakpoint cloning procedure in a case of renal cell cancer (RCC), we found that the FBXW7 gene was disrupted by a constitutional t(3;4)(q21;q31). Subsequent analysis of the tumor tissue revealed the presence of several anomalies, including loss of the derivative chromosome 3. Upon screening of a cohort of 29 independent primary RCCs, we identified one novel pathogenic mutation, suggesting that the FBXW7 gene may also play a role in the development of sporadic RCCs. In addition, we screened a cohort of 48 unrelated familial RCC cases with unknown etiology. Except for several known or benign sequence variants such as single nucleotide polymorphisms (SNPs), no additional pathogenic variants were found. Previous mouse models have suggested that the FBXW7 gene may play a role in the predisposition to tumor development. Here we report that disruption of this gene may predispose to the development of human RCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Cell Cycle Proteins/genetics , Chromosomes, Human, Pair 3 , Chromosomes, Human, Pair 4 , F-Box Proteins/genetics , Kidney Neoplasms/genetics , Translocation, Genetic , Ubiquitin-Protein Ligases/genetics , Base Sequence , DNA Primers , F-Box-WD Repeat-Containing Protein 7 , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Mutation , Polymerase Chain Reaction
7.
Cancer Genet Cytogenet ; 193(1): 54-62, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19602464

ABSTRACT

Uterine leiomyomas are the most common neoplasms in women of reproductive age. Approximately 40% of these neoplasms show recurring structural cytogenetic anomalies, including del(7)(q22), t(12;14)(q15;q24), t(1;2)(p36;p24), and anomalies affecting 6p21 or 10q22. Using positional cloning strategies, we and others had previously identified HMGA1, HMGA2, RAD51L1, and MYST4 (previously referred to as MORF); as primary target (fusion) genes associated with tumor development in three of these distinct cytogenetic subgroups. Here, we report the positional cloning of a single, recurrent, leiomyoma-associated anomaly, t(1;2)(p36;p24). Molecular characterization of the reciprocal breakpoint intervals showed that that AJAP1 (alias SHREW1) and NPHP4 flank the breakpoint on chromosome 1 and that ITSN2 and NCOA1 flank the breakpoint on chromosome 2. Detailed analysis of the breakpoint regions revealed that in this particular case the translocation was associated with a 27-bp deletion on chromosome 1 and a 136-bp duplication on chromosome 2. No breakpoint-spanning (fusion) genes were identified. In silico prediction of transcription factor binding sites, however, indicated the presence of several such sites in the respective breakpoint regions, and major changes therein as a result of the t(1;2)(p36;p24) under investigation. We postulate that transcriptional deregulation of one or more of these breakpoint-flanking genes may contribute to the development of human uterine leiomyomas.


Subject(s)
Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 2/genetics , Leiomyoma/genetics , Translocation, Genetic , Uterine Neoplasms/genetics , Base Sequence , Binding Sites , Chromosome Aberrations , Chromosome Breakage , Chromosome Deletion , Chromosome Mapping , Cloning, Molecular , Cytogenetic Analysis , Female , Humans , Karyotyping , Molecular Sequence Data , Polymerase Chain Reaction , Transcription Factors/metabolism
8.
Br J Haematol ; 143(2): 210-21, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18699851

ABSTRACT

Mantle cell lymphoma (MCL) is characterized by the t(11;14)(q13;q32) translocation and several other cytogenetic aberrations, including heterozygous loss of chromosomal arms 1p, 6q, 11q and 13q and/or gains of 3q and 8q. The common intervals of chromosomal imbalance have been narrowed down using array-comparative genomic hybridization (CGH). However, the chromosomal intervals still contain many genes potentially involved in MCL pathogeny. Combined analysis of tiling-resolution array-CGH with gene expression profiling on 11 MCL tumours enabled the identification of genomic alterations and their corresponding gene expression profiles. Only subsets of genes located within given cytogenetic anomaly-intervals showed a concomitant change in mRNA expression level. The genes that showed consistent correlation between DNA copy number and RNA expression levels are likely to be important in MCL pathology. Besides several 'anonymous genes', we also identified various fully annotated genes, whose gene products are involved in cyclic adenosine monophosphate-regulated pathways (PRKACB), DNA damage repair, maintenance of chromosome stability and prevention of rereplication (ATM, ERCC5, FBXO5), energy metabolism (such as genes that are involved in the synthesis of proteins encoded by the mitochondrial genome) and signal transduction (ARHGAP29). Deregulation of these gene products may interfere with the signalling pathways that are involved in MCL tumour development and maintenance.


Subject(s)
Gene Dosage , Gene Expression Profiling/methods , Lymphoma, Mantle-Cell/genetics , Oligonucleotide Array Sequence Analysis/methods , Aged , Comparative Genomic Hybridization/methods , Female , Gene Amplification , Gene Deletion , Humans , Male , MicroRNAs/analysis , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction/methods , Translocation, Genetic
9.
Cancer Genet Cytogenet ; 179(1): 11-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17981209

ABSTRACT

Our group and others had previously developed a high throughput procedure to map translocation breakpoints using chromosome flow sorting in conjunction with microarray-based comparative genomic hybridization (arrayCGH). Here we applied both conventional positional cloning and integrated arrayCGH procedures to the mapping of constitutional chromosome anomalies in four patients with renal cell cancer (RCC), three with a chromosome 3 translocation, and one with an insertion involving chromosome 3. In one of these patients, who was carrying a t(3;4)(p13;p15), the KCNIP4 gene was found to be disrupted. KCNIP4 belongs to a family of potassium channel-interacting proteins and is highly expressed in normal kidney cells. In addition, KCNIP4 splice variants have specifically been encountered in RCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Kv Channel-Interacting Proteins/genetics , Translocation, Genetic , Cell Line, Tumor , Chromosome Breakage , Chromosome Mapping , Chromosomes, Human, Pair 3 , Chromosomes, Human, Pair 4 , Cloning, Molecular , Humans , In Situ Hybridization, Fluorescence , Loss of Heterozygosity , Mutagenesis, Insertional
10.
J Clin Oncol ; 24(33): 5223-33, 2006 Nov 20.
Article in English | MEDLINE | ID: mdl-17114655

ABSTRACT

PURPOSE: To delineate clinically relevant molecular signatures of intracranial ependymoma. MATERIALS AND METHODS: We analyzed 24 primary intracranial ependymomas. For genomic profiling, microarray-based comparative genomic hybridization (CGH) was used and results were validated by fluorescent in situ hybridization and loss of heterozygosity mapping. We performed gene expression profiling using microarrays, real-time quantitative reverse transcriptase polymerase chain reaction, and methylation analysis of selected genes. We applied class comparison analyses to compare both genomic and expression profiling data with clinical characteristics. RESULTS: A variable number of genomic imbalances were detected by array CGH, revealing multiple regions of recurrent gain (including 2q23, 7p21, 12p, 13q21.1, and 20p12) and loss (including 5q31, 6q26, 7q36, 15q21.1, 16q24, 17p13.3, 19p13.2, and 22q13.3). An ependymoma-specific gene expression signature was characterized by the concurrent abnormal expression of developmental and differentiation pathways, including NOTCH and sonic hedgehog signaling. We identified specific differentially imbalanced genomic clones and gene expression signatures significantly associated with tumor location, patient age at disease onset, and retrospective risk for relapse. Integrated genomic and expression profiling allowed us to identify genes of which the expression is deregulated in intracranial ependymoma, such as overexpression of the putative proto-oncogene YAP1 (located at 11q22) and downregulation of the SULT4A1 gene (at 22q13.3). CONCLUSION: The present exploratory molecular profiling study allowed us to refine previously reported intervals of genomic imbalance, to identify novel restricted regions of gain and loss, and to identify molecular signatures correlating with various clinical variables. Validation of these results on independent data sets represents the next step before translation into the clinical setting.


Subject(s)
Brain Neoplasms/genetics , Ependymoma/genetics , Gene Expression Regulation, Neoplastic , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Adult , Child , Child, Preschool , DNA Methylation , Down-Regulation , Female , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Infant , Loss of Heterozygosity , Male , Oligonucleotide Array Sequence Analysis , Phosphoproteins/genetics , Proto-Oncogene Mas , Reverse Transcriptase Polymerase Chain Reaction , Sulfotransferases/genetics , Transcription Factors , Up-Regulation , YAP-Signaling Proteins
13.
Am J Hum Genet ; 77(4): 606-16, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16175506

ABSTRACT

Mental retardation (MR) occurs in 2%-3% of the general population. Conventional karyotyping has a resolution of 5-10 million bases and detects chromosomal alterations in approximately 5% of individuals with unexplained MR. The frequency of smaller submicroscopic chromosomal alterations in these patients is unknown. Novel molecular karyotyping methods, such as array-based comparative genomic hybridization (array CGH), can detect submicroscopic chromosome alterations at a resolution of 100 kb. In this study, 100 patients with unexplained MR were analyzed using array CGH for DNA copy-number changes by use of a novel tiling-resolution genomewide microarray containing 32,447 bacterial artificial clones. Alterations were validated by fluorescence in situ hybridization and/or multiplex ligation-dependent probe amplification, and parents were tested to determine de novo occurrence. Reproducible DNA copy-number changes were present in 97% of patients. The majority of these alterations were inherited from phenotypically normal parents, which reflects normal large-scale copy-number variation. In 10% of the patients, de novo alterations considered to be clinically relevant were found: seven deletions and three duplications. These alterations varied in size from 540 kb to 12 Mb and were scattered throughout the genome. Our results indicate that the diagnostic yield of this approach in the general population of patients with MR is at least twice as high as that of standard GTG-banded karyotyping.


Subject(s)
Gene Expression Profiling , Genome, Human , Intellectual Disability/genetics , Adolescent , Adult , Child , Female , Humans , Karyotyping , Male , Nucleic Acid Hybridization
14.
Cancer Genet Cytogenet ; 161(2): 140-5, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16102584

ABSTRACT

Glioblastomas, the most frequent and malignant glial tumors, are known to be phenotypically heterogeneous. A low fraction of glioblastomas is associated with specific chromosomal losses at 1p and 19q, which are commonly found in oligodendrogliomas and are generally considered to be a primary event in the development of these tumors. Subsequent progression of oligodendroglial tumors appears to be triggered by additional molecular features underlying the transition to anaplastic oligodendroglioma and glioblastoma multiforme (GBM) such as deletions of 9p and 10q, and alterations of CDKN2A (p16), which is located at 9p21. These findings strengthen the view that GBM on rare occasions may develop from oligodendroglial differentiated cells. In the present study, we evaluated the newly established MI-4 glioblastoma cell line, which displays 1p and 19q specific alterations targeting preferential regions of allelic loss in glial neoplasms, by array-CGH and fluorescence in situ hybridization (FISH) analyses that were combined to obtain a high resolution map of targeted chromosome rearrangements and copy number changes throughout the genome. Genome-wide and chromosome 19 full coverage array-CGH analysis of the MI-4 cell line revealed that in this particular cell line, 1p-specific loss, including the CDKN2 (p18) gene, is not accompanied by loss of the previously described 19q13.3 tumor suppressor candidate region. Interestingly, the array-CGH (CGHa) profile showed an increase in copy number along most of 19q including the AKT2 oncogene and the KLKs gene family, which have previously been shown to be amplified in pancreatic carcinomas and upregulated in several tumors, respectively. The concomitant 1p partial loss and chromosome 19 alterations, with the +7 and -10-specific GBM markers associated with homozygous deletion of 9p21.3 including CDKN2A (p16), are distinct features of the glioblastoma MI-4 cell line, illustrating its origin from an olidodendroglial tumor. Based on these results, we conclude that the MI-4 glioblastoma cell line might function as a model system for investigations into the behavior of a defined oligodendroglioma subtype.


Subject(s)
Brain Neoplasms/genetics , Cell Line, Tumor , Chromosome Aberrations , Chromosomes, Human, Pair 1 , Glioblastoma/genetics , Oligodendroglioma/genetics , Chromosomes, Human, Pair 19 , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Microarray Analysis
15.
Hum Mol Genet ; 14(14): 1955-63, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15917269

ABSTRACT

Recently, we identified a patient with an infantile sacrococcygeal teratoma and a constitutional t(12;15)(q13;q25). Here, we show that, as a result of this chromosomal translocation, the SUMO/Sentrin-specific protease 1 gene (SENP1) on chromosome 12 and the embryonic polarity-related mesoderm development gene (MESDC2) on chromosome 15 are disrupted and fused. Both reciprocal SENP1-MESDC2 (SEME) and MESDC2-SENP1 (MESE) fusion genes are transcribed in tumor-derived cells and their open reading frames encode aberrant proteins. As a consequence of this, and in contrast to wild-type (WT) MESDC2, the translocation-associated SEME protein is no longer targeted to the endoplasmatic reticulum, leading to a presumed loss-of-function as a chaperone for the WNT co-receptors LRP5 and/or LRP6. Ultimately, this might lead to abnormal development and/or routing of germ cell tumor precursor cells. SUMO, a post-translational modifier, plays an important role in several cellular key processes and is cleaved from its substrates by WT SENP1. Using a PML desumoylation assay, we found that translocation-associated MESE proteins exhibit desumoylation capacities similar to those observed for WT SENP1. We speculate that spatio-temporal disturbances in desumoylating activities during critical stages of embryonic development might have predisposed the patient. Together, the constitutional t(12;15)(q13;q25) translocation revealed two novel candidate genes for neonatal/infantile GCT development: MESDC2 and SENP1.


Subject(s)
Cell Polarity , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 15 , Endopeptidases/genetics , Molecular Chaperones/genetics , Teratoma/genetics , Translocation, Genetic , Animals , Blotting, Southern , Cell Line , Cricetinae , Cysteine Endopeptidases , DNA Primers , Humans , In Situ Hybridization, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions/metabolism
16.
Oncogene ; 24(10): 1794-801, 2005 Mar 03.
Article in English | MEDLINE | ID: mdl-15688027

ABSTRACT

DNA copy number alterations are believed to play a major role in the development and progression of human neoplasms. Although most of these genomic imbalances have been associated with dysregulation of individual genes, their large-scale transcriptional consequences remain unclear. Pancreatic carcinomas frequently display gene copy number variation of entire chromosomes as well as of chromosomal subregions. These changes range from homozygous deletions to high-level amplifications and are believed to constitute key genetic alterations in the cellular transformation of this tumor type. To investigate the transcriptional consequences of the most drastic genomic changes, that is, genomic amplifications, and to analyse the genome-wide transcriptional effects of DNA copy number changes, we performed expression profiling of 29 pancreatic carcinoma cell lines and compared the results with matching genomic profiling data. We show that a strong association between DNA copy numbers and mRNA expression levels is present in pancreatic cancer, and demonstrate that as much as 60% of the genes within highly amplified genomic regions display associated overexpression. Consequently, we identified 67 recurrently overexpressed genes located in seven precisely mapped commonly amplified regions. The presented findings indicate that more than one putative target gene may be of importance in most pancreatic cancer amplicons.


Subject(s)
Gene Amplification , Gene Dosage , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/genetics , Chromosome Mapping , Humans
17.
Blood ; 105(4): 1686-93, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15498857

ABSTRACT

Mantle cell lymphoma (MCL) is an aggressive, highly proliferative B-cell non-Hodgkin lymphoma, characterized by the specific t(11;14)(q13;q32) translocation. It is well established that this translocation alone is not sufficient to promote MCL development, but that additional genetic changes are essential for malignant transformation. We have identified such additional tumorigenic triggers in MCL tumors, by applying genome-wide array-based comparative genomic hybridization with an 800-kilobase (kb) resolution. This strategy, combined with a newly developed statistical approach, enabled us to confirm previously reported genomic alterations such as loss of 1p, 6q, 11q, 13q and gain of 3q and 8q, but it also facilitated the detection of novel recurrent genomic imbalances, such as gain of 4p12-13 and loss of 20p12.1-12.3, 20q12-13.2, 22q12.1-12.3, and 22q13.31-13.32. Genomic hotspot detection allowed for the identification of small genomic intervals that are frequently affected (57%-93%), resulting in interesting positional candidate genes such as KITLG, GPC5, and ING1. Finally, by assessing multiple biopsies from the same patient, we show that seemingly stable genomes do show subtle genomic changes over time. The follow-up of multiple biopsies of patients with MCL by high-resolution genomic profiling is expected to provide us with new clues regarding the relation between clinical outcome and in vivo cytogenetic evolution.


Subject(s)
Chromosome Aberrations , Genome, Human , Lymphoma, Mantle-Cell/genetics , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Aged , Aged, 80 and over , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 20/genetics , Chromosomes, Human, Pair 22/genetics , Chromosomes, Human, Pair 4/genetics , Disease Progression , Female , Humans , Male , Middle Aged , Neoplasm Recurrence, Local , Nucleic Acid Hybridization/methods , Oligonucleotide Array Sequence Analysis/methods
18.
Curr Mol Med ; 4(8): 849-54, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15579032

ABSTRACT

Renal cell carcinomas (RCCs) occur in both sporadic and familial forms. In a subset of families the occurrence of RCCs co-segregates with the presence of constitutional chromosome 3 translocations. Previously, such co-segregation phenomena have been widely employed to identify candidate genes in various hereditary (cancer) syndromes. Here we survey the translocation 3-positive RCC families that have been reported to date and the subsequent identification of its respective candidate genes using positional cloning strategies. Based on allele segregation, loss of heterozygosity and mutation analyses of the tumors, a multi-step model for familial RCC development has been generated. This model is relevant for (i) understanding familial tumorigenesis and (ii) rational patient management. In addition, a high throughput microarray-based strategy is presented that will enable the rapid identification of novel positional candidate genes via a single step procedure. The functional consequences of the (fusion) genes that have been identified so far, the multi-step model and its consequences for clinical diagnosis, the identification of persons at risk and genetic counseling in RCC families are discussed.


Subject(s)
Carcinoma, Renal Cell/genetics , Chromosomes, Human, Pair 3 , Kidney Neoplasms/genetics , Translocation, Genetic , Adult , Aged , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 2 , Family , Genetic Counseling , Humans , Middle Aged , Oligonucleotide Array Sequence Analysis
19.
Nat Genet ; 36(9): 955-7, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15300250

ABSTRACT

CHARGE syndrome is a common cause of congenital anomalies affecting several tissues in a nonrandom fashion. We report a 2.3-Mb de novo overlapping microdeletion on chromosome 8q12 identified by array comparative genomic hybridization in two individuals with CHARGE syndrome. Sequence analysis of genes located in this region detected mutations in the gene CHD7 in 10 of 17 individuals with CHARGE syndrome without microdeletions, accounting for the disease in most affected individuals.


Subject(s)
Abnormalities, Multiple/genetics , Choanal Atresia/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Heart Defects, Congenital/genetics , Mutation , Coloboma/genetics , Deafness/genetics , Gene Deletion , Humans , Sequence Analysis, DNA , Syndrome
20.
Nucleic Acids Res ; 32(8): 2315-22, 2004.
Article in English | MEDLINE | ID: mdl-15118077

ABSTRACT

The MiTF/TFE (MiT) family of basic helix-loop-helix leucine zipper transcription factors is composed of four closely related members, MiTF, TFE3, TFEB and TFEC, which can bind target DNA both as homo- or heterodimers. Using real-time RT-PCR, we have analyzed the relative expression levels of the four members in a broad range of human tissues, and found that their ratio of expression is tissue-dependent. We found that, similar to the MiTF gene, the genes for TFEB and TFEC contain multiple alternative first exons with restricted and differential tissue distributions. Seven alternative 5' exons were identified in the TFEB gene, of which three displayed specific expression in placenta and brain, respectively. A novel TFEC transcript (TFEC-C) encodes an N-terminally truncated TFEC isoform lacking the acidic activation domain (AAD), and is exclusively expressed in kidney and small intestine. Furthermore, we observed that a considerable proportion of the TFEC transcripts splice out protein-coding exons, resulting in transcription factor isoforms lacking one or more functional domains, primarily the basic region and/or the AAD. These isoforms were always co-expressed with the intact transcription factors and may act as negative regulators of MiTF/TFE proteins. Our data reveal that multiple levels of regulation exist for the MiTF/TFE family of transcription factors, which indicates how these transcription factors may participate in various cellular processes in different tissues.


Subject(s)
Alternative Splicing , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Neoplasm Proteins , Transcription Factors/chemistry , Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , DNA-Binding Proteins/metabolism , Exons , Gene Expression , Helix-Loop-Helix Motifs , Humans , Leucine Zippers , Microphthalmia-Associated Transcription Factor , Protein Structure, Tertiary , RNA, Messenger/metabolism , Tissue Distribution , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...