Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Acta Neuropathol ; 126(2): 291-301, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23660940

ABSTRACT

Pilocytic astrocytomas (PAs) are the most common brain tumors in pediatric patients and can cause significant morbidity, including chronic neurological deficiencies. They are characterized by activating alterations in the mitogen-activated protein kinase pathway, but little else is known about their development. To map the global DNA methylation profiles of these tumors, we analyzed 62 PAs and 7 normal cerebellum samples using Illumina 450K microarrays. These data revealed two subgroups of PA that separate according to tumor location (infratentorial versus supratentorial), and identified key neural developmental genes that are differentially methylated between the two groups, including NR2E1 and EN2. Integration with transcriptome microarray data highlighted significant expression differences, which were unexpectedly associated with a strong positive correlation between methylation and expression. Differentially methylated probes were often identified within the gene body and/or regions up- or downstream of the gene, rather than at the transcription start site. We also identified a large number of differentially methylated genes between cerebellar PAs and normal cerebellum, which were again enriched for developmental genes. In addition, we found a significant association between differentially methylated genes and SUZ12 binding sites, indicating potential disruption of the polycomb repressor complex 2 (PRC2). Taken together, these data suggest that PA from different locations in the brain may arise from region-specific cells of origin, and highlight the potential disruption of key developmental regulators during tumorigenesis. These findings have implications for future basic research and clinical trials, as therapeutic targets and drug sensitivity may differ according to tumor location.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Cerebellar Neoplasms/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Astrocytoma/pathology , Binding Sites/genetics , Brain Neoplasms/pathology , Cerebellar Neoplasms/pathology , Child , DNA Methylation/genetics , Gene Expression Profiling , Genes, Developmental/genetics , Humans , Neoplasm Proteins , Oligonucleotide Array Sequence Analysis , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Transcription Factors
2.
Int J Cancer ; 131(5): 1104-13, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22020830

ABSTRACT

We have previously identified a region containing 16 CpGs within the MGMT CpG islands which is critical for the transcriptional control of MGMT (Malley, Acta Neuropathol 2011). To investigate the patterns and incidence of MGMT methylation in astrocytic and oligodendroglial tumors, we quantitatively assessed methylation at these 16 CpGs using bisulfite modification followed by pyrosequencing of 362 gliomas not treated with temozolomide, and correlated the findings with previously identified patterns of genetic abnormalities, patients' age and survival. The MGMT gene was considered to be methylated when the mean methylation of the 16 CpGs was 10% or higher. This cut-off value distinguished diffuse astrocytomas with high and low MGMT expression. Within each tumor type, the patterns of methylation were highly variable and also highly heterogeneous across the 16 CpGs. A high incidence of MGMT methylation was observed in all subtypes of gliomas included in this study. Among a subset of 97 tumors where conventional methylation-specific PCR (MSP) was also applied, methylation was detected by both methods in 54 tumors, while the pyrosequencing results identified a further 17 tumors. No additional cases were found using MSP alone, indicating that pyrosequencing is a robust method for methylation analysis. All tumors with IDH1/IDH2 mutations except two had MGMT methylation, while there were many tumors with MGMT methylation, particularly primary glioblastomas, which had no mutations of IDH1/2. We suggest that MGMT methylation may be one of the earliest events in the development of astrocytic and oligodendroglial tumors.


Subject(s)
Astrocytoma/genetics , CpG Islands/genetics , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Oligodendroglioma/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Astrocytoma/mortality , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Child , DNA, Neoplasm/genetics , Female , Humans , Male , Middle Aged , Oligodendroglioma/mortality , Prognosis , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction , Survival Rate , Young Adult
3.
Acta Neuropathol ; 121(6): 753-61, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21327941

ABSTRACT

Diffuse astrocytomas (WHO grade II) typically present as slow-growing tumours showing significant cellular differentiation, but possessing a tendency towards malignant progression. They account for ~10% of all astrocytic tumours, with a peak incidence between 30 and 40 years of age. Median survival is reported as around 6-8 years. Mutations of TP53 and IDH1 have been described as genetic hallmarks, while copy number alterations are also relatively common. However, there is some evidence to suggest that these characteristics may vary with age. Here, we present an integrated clinicopathologic, genomic and transcriptomic analysis suggesting that paediatric and adult tumours are associated with distinct genetic signatures. For example, no childhood tumour showed mutation of IDH1/2 or TP53, virtually no copy number changes were seen, and MGMT methylation was absent. In contrast, adult tumours showed IDH1/2 mutation in 94% and TP53 mutation in 69% of cases, with multiple copy number alterations per case and hypermethylation of MGMT in the majority of tumours. These differences were associated with a worse prognosis in the adult patients. The expression array data also revealed a significant difference in the expression of a number of genes putatively involved in neural stem cell maintenance and CNS development, including DLL3, HES5, BMP2, TIMP1 and BAMBI. Genes involved in DNA replication and the cell cycle were also enriched in the adult tumours, suggesting that their more aggressive behaviour may be due to derivation from a more rapidly dividing, less differentiated cell type.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Gene Expression Regulation, Neoplastic/genetics , Isocitrate Dehydrogenase/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Age Factors , Astrocytoma/pathology , Astrocytoma/physiopathology , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Child , DNA Copy Number Variations , DNA Methylation , DNA Mutational Analysis/methods , Female , Gene Expression Profiling/methods , Humans , Male , Middle Aged , Mutation/genetics , Oligonucleotide Array Sequence Analysis/methods , Principal Component Analysis , Survival Analysis , Young Adult
4.
Acta Neuropathol ; 121(5): 651-61, 2011 May.
Article in English | MEDLINE | ID: mdl-21287394

ABSTRACT

O(6)-Methylguanine-DNA methyltransferase (MGMT) is a DNA repair protein that removes alkyl DNA adducts such as those induced by alkylating agents. Loss of MGMT expression through transcriptional silencing by hypermethylation of its CpG island (CGI) is found in diverse human cancers including glioblastomas. Glioblastomas that have MGMT methylation respond to temozolomide, an alkylating agent, resulting in improved survival. Consequently, assessment of MGMT methylation has become a therapy response and prognostic indicator. However, it is not clear whether the region of the MGMT CGI commonly analysed is the critical region involved in transcriptional control. We measured methylation levels at each CpG site for the entire MGMT CGI using bisulfite modification and pyrosequencing, and compared them with MGMT mRNA expression in glioblastoma cell lines, xenografts and normal brain tissues (41 samples). Two critical regions were identified (DMR1 and DMR2). DMR2 encompasses the commonly analysed region and was always methylated when DMR1 was methylated. A luciferase reporter assay showed that substitutions of several specific CpG sites within DMR2 significantly attenuated the promoter activity of the MGMT CGI. Our results indicate that several CpG sites within DMR2 play a critical role in the transcriptional control of MGMT, making DMR2 the optimal target for methylation testing. However, given the highly variable patterns of MGMT methylation associated with transcriptional silencing observed in this region among the tumours in this study, methylation levels need to be measured at a number of individual CpGs within DMR2 to confidently predict transcriptional silencing and thus sensitivity to alkylating agents.


Subject(s)
Brain Neoplasms/genetics , CpG Islands/genetics , DNA Methylation/physiology , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Glioblastoma/genetics , Transcription, Genetic/physiology , Tumor Suppressor Proteins/genetics , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cell Line, Tumor , DNA Methylation/drug effects , DNA Modification Methylases/chemistry , DNA Modification Methylases/metabolism , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , Drug Delivery Systems/methods , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Transcription, Genetic/drug effects , Transplantation, Heterologous/pathology , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism
5.
Neuro Oncol ; 12(7): 664-78, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20164239

ABSTRACT

We studied the status of chromosomes 1 and 19 in 363 astrocytic and oligodendroglial tumors. Whereas the predominant pattern of copy number abnormality was a concurrent loss of the entire 1p and 19q regions (total 1p/19q loss) among oligodendroglial tumors and partial deletions of 1p and/or 19q in astrocytic tumors, a subset of apparently astrocytic tumors also had total 1p/19q loss. The presence of total 1p/19q loss was associated with longer survival of patients with all types of adult gliomas independent of age and diagnosis (P = .041). The most commonly deleted region on 19q in astrocytic tumors spans 885 kb in 19q13.33-q13.41, which is telomeric to the previously proposed region. Novel regions of homozygous deletion, including a part of DPYD (1p21.3) or the KLK cluster (19q13.33), were observed in anaplastic oligodendrogliomas. Amplifications encompassing AKT2 (19q13.2) or CCNE1 (19q12) were identified in some glioblastomas. Deletion mapping of the centromeric regions of 1p and 19q in the tumors that had total 1p/19q loss, indicating that the breakpoints lie centromeric to NOTCH2 within the pericentromeric regions of 1p and 19q. Thus, we show that the copy number abnormalities of 1p and 19q in human gliomas are complex and have distinct patterns that are prognostically predictive independent of age and pathological diagnosis. An accurate identification of total 1p/19q loss and discriminating this from other 1p/19q changes is, however, critical when the 1p/19q copy number status is used to stratify patients in clinical trials.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Chromosome Deletion , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 1/genetics , Glioma/diagnosis , Glioma/genetics , Brain Neoplasms/classification , Chromosome Aberrations , Glioma/classification , Humans , Prognosis
6.
Neuro Oncol ; 11(4): 341-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19435942

ABSTRACT

We screened exon 4 of the gene isocitrate dehydrogenase 1 (NADP+), soluble (IDH1) for mutations in 596 primary intracranial tumors of all major types. Codon 132 mutation was seen in 54% of astrocytomas and 65% of oligodendroglial tumors but in only 6% of glioblastomas (3% of primary and 50% of secondary glioblastomas). There were no mutations in any other type of tumor studied. While mutations in the tumor protein p53 gene (TP53) and total 1p/19q deletions were mutually exclusive, IDH1 mutations were strongly correlated with these genetic abnormalities. All four types of mutant IDH1 proteins showed decreased enzymatic activity. The data indicate that IDH1 mutation combined with either TP53 mutation or total 1p/19q loss is a frequent and early change in the majority of oligodendroglial tumors, diffuse astrocytomas, anaplastic astrocytomas, and secondary glioblastomas but not in primary glioblastomas.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Oligodendroglioma/genetics , Adult , Biomarkers, Tumor/genetics , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 19/genetics , Comparative Genomic Hybridization , Exons/genetics , Genotype , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Loss of Heterozygosity , Oligodendroglioma/enzymology , Oligodendroglioma/pathology , Prognosis , Tumor Suppressor Protein p53/genetics
7.
Genes Chromosomes Cancer ; 48(2): 121-31, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18973140

ABSTRACT

Isodicentric 17q is the most commonly reported chromosomal abnormality in medulloblastomas. Its frequency suggests that genes disrupted in medulloblastoma formation may play a role in tumorigenesis. We have previously identified two chromosome 17 breakpoint at a 1 Mb resolution. Our aims were to accurately map the position of these breakpoints and to identify mechanisms of gene disruption at this site. CGH with a custom tiling path genomic BAC array of chromosome 17 enriched with fosmids at the breakpoint regions was used to analyze a series of 45 medulloblastomas and three medulloblastoma-derived cell lines. In total, 17 of 45 medulloblastomas had an isodicentric 17q. Two breakpoint regions were identified and their positions were mapped. The array identified a more complex arrangement at the breakpoint than has been reported previously using lower resolution BAC arrays. The patterns observed indicated that dicentric chromosome formation occurs both via nonallelic homologous recombination between palindromically arranged low copy repeats (the previously accepted mechanism) and by recombination between nonidentical sequences. In addition, novel alternative structural alterations, a homozygous deletion and a duplication, were identified within the chromosome breakpoint region in two cases. At the resolution of the array, these structural alterations spanned the same genes as cases with dicentric 17q formation, implying that the disruption of genes at the chromosome breakpoint itself may be of greater biological significance than has previously been suspected.


Subject(s)
Chromosome Breakage , Chromosomes, Human, Pair 17/genetics , Medulloblastoma/genetics , Adolescent , Adult , Child , Child, Preschool , Chromosome Mapping , Comparative Genomic Hybridization , Female , Gene Deletion , Gene Dosage , Gene Duplication , Humans , Male , Oligonucleotide Array Sequence Analysis , Recombination, Genetic
8.
Cancer Res ; 68(21): 8673-7, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18974108

ABSTRACT

Brain tumors are the most common solid tumors of childhood, and pilocytic astrocytomas (PA) are the most common central nervous system tumor in 5 to 19 year olds. Little is known about the genetic alterations underlying their development. Here, we describe a tandem duplication of approximately 2 Mb at 7q34 occurring in 66% of PAs. This rearrangement, which was not observed in a series of 244 higher-grade astrocytomas, results in an in-frame fusion gene incorporating the kinase domain of the BRAF oncogene. We further show that the resulting fusion protein has constitutive BRAF kinase activity and is able to transform NIH3T3 cells. This is the first report of BRAF activation through rearrangement as a frequent feature in a sporadic tumor. The frequency and specificity of this change underline its potential both as a therapeutic target and as a diagnostic tool.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Gene Duplication , Gene Fusion , Proto-Oncogene Proteins B-raf/genetics , Animals , COS Cells , Chlorocebus aethiops , Chromosomes, Human, Pair 7 , Genes, ras , Humans , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Promoter Regions, Genetic , Survival Analysis
9.
Brain Pathol ; 18(4): 469-73, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18397339

ABSTRACT

Subependymomas (SE) are slow-growing brain tumors that tend to occur within the ventricles of middle-aged and elderly adults. The World Health Organization classifies these tumors within the ependymoma group. Previous limited analysis of this tumor type had not revealed significant underlying cytogenetic abnormalities. We have used microarray comparative genomic hybridization to study a series of SE (n = 12). A whole-genome array at 0.97-Mb resolution showed copy number abnormalities in five of 12 cases (42%). Two cases (17%) showed regions of loss on chromosome 6. More detailed analysis of all cases using a chromosome 6 tile-path array confirmed the presence of overlapping regions of loss in only these two cases. One of these cases also showed trisomy chromosome 7. Monosomy of chromosome 8 was seen in a further two cases (17%), and a partial loss on chromosome 14 was observed in one additional case. This is the first array-based, genome-wide study of SE. The observation that five of 12 cases examined (42%) at 0.97-Mb resolution showed chromosomal copy number abnormalities is a novel finding in this tumor type.


Subject(s)
Cerebral Ventricle Neoplasms/genetics , Chromosomes, Human/genetics , Gene Dosage/genetics , Genetic Predisposition to Disease/genetics , Glioma, Subependymal/genetics , Monosomy , Trisomy , Adult , Aged , Cerebral Ventricle Neoplasms/metabolism , Cerebral Ventricle Neoplasms/pathology , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 6/genetics , Chromosomes, Human, Pair 7/genetics , Chromosomes, Human, Pair 8/genetics , DNA Mutational Analysis , Female , Gene Expression Profiling , Genomic Library , Genotype , Glioma, Subependymal/metabolism , Glioma, Subependymal/pathology , Humans , Male , Middle Aged , Mutation/genetics , Oligonucleotide Array Sequence Analysis , Sequence Deletion
10.
J Neuropathol Exp Neurol ; 65(11): 1049-58, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17086101

ABSTRACT

Brain tumors are the most common solid tumors of childhood, accounting for over 20% of cancers in children under 15 years of age. Pilocytic astrocytomas (PAs), World Health Organization grade I, are one of the most frequently occurring childhood brain tumors, yet little is known about genetic changes characterizing this entity. We have used microarray comparative genomic hybridization at 0.97 Mb resolution to study a series of PAs (n = 44). No copy number abnormality was seen in 64% of cases at this resolution. However, whole chromosomal gain (median 5 chromosomes affected) occurred in 32% of tumors. The most frequently affected chromosomes were 5 and 7 (11 of 44 cases each) followed by 6, 11, 15, and 20 (greater than 10% of cases each). Findings were confirmed by fluorescence in situ hybridization and microsatellite analysis in a subset of tumors. Chromosomal gain was significantly more frequent in PAs from patients over 15 years old (p = 0.03, Fisher exact test). The number of chromosomes involved was also significantly greater in the older group (p = 0.02, Mann-Whitney U test). One case (2%) showed a region of gain on chromosome 3 and one (2%) a deletion on 6q as their sole abnormalities. This is the first genomewide study to show this nonrandom pattern of genetic alteration in pilocytic astrocytomas.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Gene Dosage , Oligonucleotide Array Sequence Analysis , Adolescent , Adult , Age Factors , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Astrocytoma/pathology , Brain Neoplasms/pathology , Child , Child, Preschool , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Infant , Leukocyte Common Antigens/metabolism , Male , Microglia/metabolism , Microsatellite Repeats
11.
J Neuropathol Exp Neurol ; 65(6): 549-61, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16783165

ABSTRACT

Medulloblastomas and supratentorial primitive neuroectodermal tumors are aggressive childhood tumors. We report our findings using array comparative genomic hybridization (CGH) on a whole-genome BAC/PAC/cosmid array with a median clone separation of 0.97 Mb to study 34 medulloblastomas and 7 supratentorial primitive neuroectodermal tumors. Array CGH allowed identification and mapping of numerous novel, small regions of copy number change to genomic sequence in addition to the large regions already known from previous studies. Novel amplifications were identified, some encompassing oncogenes MYCL1, PDGFRA, KIT, and MYB not previously reported to show amplification in these tumors. In addition, one supratentorial primitive neuroectodermal tumor had lost both copies of the tumor-suppressor genes CDKN2A and CDKN2B. Ten medulloblastomas had findings suggestive of isochromosome 17q. In contrast to previous reports using conventional CGH, array CGH identified 3 distinct breakpoints in these cases: Ch 17: 17940393-19251679 (17p11.2, n = 6), Ch 17: 20111990-23308272 (17p11.2-17q11.2, n = 4), and Ch 17: 38425359-39091575 (17q21.31, n = 1). Significant differences were found in the patterns of copy number change between medulloblastomas and supratentorial primitive neuroectodermal tumors, providing further evidence that these tumors are genetically distinct despite their morphologic and behavioral similarities.


Subject(s)
Brain Neoplasms/genetics , Medulloblastoma/genetics , Neuroectodermal Tumors, Primitive/genetics , Nucleic Acid Hybridization/methods , Supratentorial Neoplasms/genetics , Adolescent , Adult , Child , Child, Preschool , Chromosome Aberrations , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence/methods , Infant , Reverse Transcriptase Polymerase Chain Reaction/methods
12.
Lab Invest ; 86(9): 968-78, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16751780

ABSTRACT

Array-based comparative genomic hybridisation (aCGH) has diverse applications in cancer gene discovery and translational research. Currently, aCGH is performed primarily using high molecular weight DNA samples and its application to formalin-fixed and paraffin-embedded (FFPE) tissues remains to be established. To explore how aCGH can be reliably applied to archival FFPE tissues and whether it is possible to apply aCGH to small numbers of cells microdissected from FFPE tissue sections, we have systematically performed aCGH on 15 pairs of matched frozen and FFPE astrocytic tumour tissues using a well-established in-house human 1 Mb BAC/PAC genomic array. By spiking tumour DNA with normal DNA, we demonstrated that at least 70% of tumour DNA was required for reliable aCGH analysis. Using aCGH data from frozen tissue as a reference, it was found that only FFPE astrocytic tumour tissues that supported PCR amplification of >300 bp DNA fragment provided high quality, reproducible aCGH data. The presence of necrosis in a tissue specimen had an adverse effect on the quality of aCGH, while fixation in formalin for up to 96 h of fresh tissue did not appear to affect the quality of the result. As little as 10-20 ng DNA from frozen or FFPE tissues could be readily used for aCGH analysis following whole genome amplification (WGA). Furthermore, as few as 2000 microdissected cells from haematoxylin-stained slides of archival FFPE tissues could be successfully used for aCGH investigations when WGA was used. By careful assessment of DNA integrity and review of histology, to exclude necrosis and select specimens with a high proportion of tumour cells, it is feasible to preselect archival FFPE tissues adequate for aCGH analysis. With the help of microdissection and WGA, it is also possible to apply aCGH to histologically defined lesions, such as carcinoma in situ.


Subject(s)
Astrocytoma/metabolism , Biomarkers, Tumor/metabolism , DNA, Neoplasm/analysis , Oligonucleotide Array Sequence Analysis/methods , Astrocytoma/pathology , Biological Specimen Banks , Formaldehyde , Humans , Microdissection , Necrosis , Paraffin Embedding , Reproducibility of Results , Time Factors , Tissue Preservation
13.
Genes Chromosomes Cancer ; 43(2): 181-93, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15770670

ABSTRACT

Many studies have reported chromosome 22 as being abnormal in astrocytic tumors. In an attempt to map precisely the abnormal region or regions that potentially harbor tumor-suppressor genes or oncogenes, we constructed a chromosome 22 tile path array covering 82% of 22q with the use of 441 chromosome 22 clones. A 10-Mb whole-genome array consisting of 270 clones from all autosomes was included in the array. A total of 126 astrocytic tumors-5 diffuse astrocytomas (A), 29 anaplastic astrocytomas (AA), and 92 glioblastomas (GB)-were examined for chromosome 22 alterations both by microsatellite analysis (using 28 markers to identify allelic imbalance) and with the tile path array. The results showed that chromosome 22 alterations in astrocytic tumors could be complex. A number of tumors had a combination of deletions with and without reduplication of the retained chromosome, as well as copy number gains and amplifications. In two glioblastomas, overlapping homozygous deletions were identified that involved three genes (DEPDC5/KIAA0645, YWHAH, C22ORF24/HSN44A4A). The terminal region telomeric to the clone RP3-398C22 appeared to be the most frequently deleted region. The estimated incidence of any chromosome 22 alteration was 5% in A, 33% in AA, and 38% in GB. This study demonstrated the advantages of combining array comparative genomic hybridization and microsatellite analysis in elucidating complex genomic rearrangements in primary human tumor tissue. Supplementary material for this article can be found on the Genes, Chromosomes and Cancer website at http://www.interscience.wiley.com/jpages/1045-2257/suppmat/index.html.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Chromosomes, Human, Pair 22 , Microsatellite Repeats/genetics , Base Sequence , DNA Primers , Humans , Nucleic Acid Hybridization , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...