Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Neuro Oncol ; 13(1): 84-98, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20926426

ABSTRACT

Extensive genomic and gene expression studies have been performed in gliomas, but the epigenetic alterations that characterize different subtypes of gliomas remain largely unknown. Here, we analyzed the methylation patterns of 807 genes (1536 CpGs) in a series of 33 low-grade gliomas (LGGs), 36 glioblastomas (GBMs), 8 paired initial and recurrent gliomas, and 9 controls. This analysis was performed with Illumina's Golden Gate Bead methylation arrays and was correlated with clinical, histological, genomic, gene expression, and genotyping data, including IDH1 mutations. Unsupervised hierarchical clustering resulted in 2 groups of gliomas: a group corresponding to de novo GBMs and a group consisting of LGGs, recurrent anaplastic gliomas, and secondary GBMs. When compared with de novo GBMs and controls, this latter group was characterized by a very high frequency of IDH1 mutations and by a hypermethylated profile similar to the recently described glioma CpG island methylator phenotype. MGMT methylation was more frequent in this group. Among the LGG cluster, 1p19q codeleted LGG displayed a distinct methylation profile. A study of paired initial and recurrent gliomas demonstrated that methylation profiles were remarkably stable across glioma evolution, even during anaplastic transformation, suggesting that epigenetic alterations occur early during gliomagenesis. Using the Cancer Genome Atlas data set, we demonstrated that GBM samples that had an LGG-like hypermethylated profile had a high rate of IDH1 mutations and a better outcome. Finally, we identified several hypermethylated and downregulated genes that may be associated with LGG and GBM oncogenesis, LGG oncogenesis, 1p19q codeleted LGG oncogenesis, and GBM oncogenesis.


Subject(s)
Brain Neoplasms/classification , Brain Neoplasms/pathology , DNA Methylation , Glioma/classification , Glioma/pathology , Adult , Aged , Aged, 80 and over , Brain Neoplasms/genetics , Cell Transformation, Neoplastic , Chromosomes, Human, Pair 1/genetics , Comparative Genomic Hybridization , CpG Islands , Epigenomics , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Glioma/genetics , Humans , Isocitrate Dehydrogenase/genetics , Middle Aged , Mutation/genetics , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Promoter Regions, Genetic/genetics , Survival Rate
2.
J Neurooncol ; 94(2): 169-72, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19255724

ABSTRACT

Expression of CHI3L1 (YKL-40) has been correlated with prognosis of glioblastoma. The variant allele (-131C-->G) of CHI3L1 promoter results in a lower transcription of CHI3L1. Therefore, we tested the hypothesis that the G variant could protect against the risk of gliomas or have a favorable prognostic impact. DNA from 296 glioblastoma patients and 190 controls were genotyped on the -131 allele. Tumor RNA was obtained from 108 patients for CHI3L1 transcript quantification. Neither genotype nor allele distribution differed between patients and controls. There was no significant difference in survival between the CC, CG, and GG patients despite the few GG patients tended to have a longer survival. There was no correlation between genotype and CHI3L1 expression in tumor samples. Taken together our data suggest that the variant allele (-131C-->G) of CHI3L1 promoter has no significant impact on survival and is not a prognostic factor for glioblastoma.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Glycoproteins/genetics , Polymorphism, Single Nucleotide/genetics , Adipokines , Adult , Aged , Aged, 80 and over , Brain Neoplasms/blood , Case-Control Studies , Chitinase-3-Like Protein 1 , DNA/blood , DNA/genetics , Female , Genotype , Glioblastoma/blood , Humans , Lectins , Male , Middle Aged , Phenotype , Prognosis , Promoter Regions, Genetic , Risk Factors , Survival Rate , Young Adult
3.
Neuro Oncol ; 11(4): 348-56, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19224763

ABSTRACT

The O(6)-methylguanine-DNA methyltransferase gene (MGMT) is methylated in several cancers, including gliomas. However, the functional role of cysteine-phosphate-guanine (CpG) island (CGI) methylation in MGMT silencing is still controversial. The aim of this study was to investigate whether MGMT CGI methylation correlates inversely with RNA expression of MGMT in glioblastomas and to determine the CpG region whose methylation best reflects the level of expression. The methylation level of CpG sites that are potentially related to expression was investigated in 54 glioblastomas by pyrosequencing, a highly quantitative method, and analyzed with respect to their MGMT mRNA expression status. Three groups of patients were identified according to the methylation pattern of all 52 analyzed CpG sites. Overall, an 85% rate of concordance was observed between methylation and expression (p < 0.0001). When analyzing each CpG separately, six CpG sites were highly correlated with expression (p < 0.0001), and two CpG regions could be used as surrogate markers for RNA expression in 81.5% of the patients. This study indicates that there is good statistical agreement between MGMT methylation and expression, and that some CpG regions better reflect MGMT expression than do others. However, if transcriptional repression is the key mechanism in explaining the higher chemosensitivity of MGMT-methylated tumors, a substantial rate of discordance should lead clinicians to be cautious when deciding on a therapeutic strategy based on MGMT methylation status alone.


Subject(s)
Brain Neoplasms/genetics , DNA Methylation , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Gene Expression Regulation, Enzymologic , Glioblastoma/genetics , Promoter Regions, Genetic/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Adolescent , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , CpG Islands , Genotype , Glioblastoma/enzymology , Glioblastoma/pathology , Humans
4.
J Neurooncol ; 90(2): 133-40, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18618226

ABSTRACT

Using a one-megabase BAC-based array comparative genomic hybridization technique (aCGH), we have investigated a series of 16 low-grade gliomas (LGGs) and their subsequent progression to higher-grade malignancies. The most frequent chromosome imbalances in primary tumors were gains of chromosomes 7q, 8q, and 22q, and losses of chromosomes 1p, 13q, and 19q. In tumor progression, gains of chromosomes 11q, 7q, 20q, and 21q, and losses of chromosomes 9p, including CDKN2A locus, 19q, 14q, 1p, and 6q were the most frequent genomic disequilibria. Progressive tumors were more imbalanced than primary tumors in terms of altered chromosomal arms (3.8 vs. 6.6 in mean abnormal chromosomal arm) and altered BACs (17 vs. 21%). Interestingly, putative novel candidate genes associated with glioma progression were identified, in particular DOCK8, PTPRD, CER1, TPHO, DHFR, MSH3, ETS1, ACACA, and CSE1L.


Subject(s)
Genome/genetics , Glioma/genetics , Glioma/metabolism , Adult , Chromosome Deletion , Chromosomes , Disease Progression , Female , Humans , Male , Microsatellite Repeats , Middle Aged , Nucleic Acid Hybridization/methods
5.
Neuro Oncol ; 10(4): 540-7, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18544654

ABSTRACT

Various gene amplifications have been observed in gliomas. Prognostic-genomic correlations testing simultaneously all these amplified genes have never been conducted in anaplastic oligodendrogliomas. A set of 38 genes that have been reported to be amplified in gliomas and investigated as the main targets of amplicons were studied in a series of 52 anaplastic oligodendrogliomas using bacterial artificial chromosome-array based comparative genomic hybridization and quantitative polymerase chain reaction. Among the 38 target genes, 15 were found to be amplified in at least one tumor. Overall, 27% of anaplastic oligodendrogliomas exhibited at least one gene amplification. The most frequently amplified genes were epidermal growth factor receptor (EGFR) and cyclin-dependent kinase 4/sarcoma amplified sequence (CDK4/SAS) in 17% and 8% of anaplastic oligodendrogliomas, respectively. Gene amplification and codeletion of chromosome arms 1p/19q were perfectly exclusive (p = 0.005). In uni- and multivariate analyses, gene amplification was a negative prognostic factor for progression-free survival and overall survival in anaplastic oligodendrogliomas, providing complementary information to the classic prognostic factors identified in anaplastic oligodendrogliomas (extent of surgery, KPS, and chromosome arms 1p/19q status).


Subject(s)
Brain Neoplasms/genetics , Gene Amplification , Oligodendroglioma/genetics , Adult , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Chromosomes, Artificial, Bacterial , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Nucleic Acid Hybridization , Oligodendroglioma/mortality , Oligodendroglioma/pathology , Prognosis , Reverse Transcriptase Polymerase Chain Reaction
6.
Mol Cancer ; 7: 41, 2008 May 20.
Article in English | MEDLINE | ID: mdl-18492260

ABSTRACT

BACKGROUND: In high grade gliomas, 1p19q codeletion and EGFR amplification are mutually exclusive and predictive of dramatically different outcomes. We performed a microarray gene expression study of four high grade gliomas with 1p19q codeletion and nine with EGFR amplification, identified by CGH-array. RESULTS: The two groups of gliomas exhibited very different gene expression profiles and were consistently distinguished by unsupervised clustering analysis. One of the most striking differences was the expression of normal brain genes by oligodendrogliomas with 1p19q codeletion. These gliomas harbored a gene expression profile that partially resembled the gene expression of normal brain samples, whereas gliomas with EGFR amplification expressed many genes in common with glioblastoma cancer stem cells. The differences between the two types of gliomas and the expression of neuronal genes in gliomas with 1p19q codeletion were both validated in an independent series of 16 gliomas using real-time RT-PCR with a set of 22 genes differentiating the two groups of gliomas (AKR1C3, ATOH8, BMP2, C20orf42, CCNB1, CDK2, CHI3L1, CTTNBP2, DCX, EGFR, GALNT13, GBP1, IGFBP2, IQGAP1, L1CAM, NCAM1, NOG, OLIG2, PDPN, PLAT, POSTN, RNF135). Immunohistochemical study of the most differentially expressed neuronal gene, alpha-internexin, clearly differentiated the two groups of gliomas, with 1p19q codeletion gliomas showing specific staining in tumor cells. CONCLUSION: These findings provide evidence for neuronal differentiation in oligodendrogliomas with 1p19q codeletion and support the hypothesis that the cell of origin for gliomas with 1p19q codeletion could be a bi-potential progenitor cell, able to give rise to both neurons and oligodendrocytes.


Subject(s)
Chromosome Deletion , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Neurons/metabolism , Oligodendroglioma/genetics , Cluster Analysis , ErbB Receptors/metabolism , Genes, Neoplasm , Genome, Human/genetics , Humans , Immunohistochemistry , Intermediate Filament Proteins/metabolism , Neoplastic Stem Cells/pathology , Oligodendroglioma/pathology , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
7.
J Neurooncol ; 88(1): 19-25, 2008 May.
Article in English | MEDLINE | ID: mdl-18253698

ABSTRACT

Immunostimulating oligonucleotides containing CpG motifs (CpG-ODN) have shown promising antitumor activity in preclinical glioma models. CpG motifs are specifically recognized by the Toll-like receptor 9 (TLR9), mainly expressed in plasmacytoid dendritic cells (pDCs) and B cells. Expression of TLR9 within human glioma samples has not been investigated. As CpG-ODN is currently under clinical trials in glioma patients, we investigated whether TLR9 is expressed at the RNA levels in a series of 37 human glioblastomas (GBM) by quantitative PCR. TLR9 expression was detected at variable levels, which might suggest that some patients are more likely to benefit from treatment with CpG-ODN than others. No significant relationships between TLR9 expression and age, sex, tumor location, lymphocytes infiltration, oligodendroglial components or survival were found. TLR9 is unlikely to be expressed by tumor cells as no TLR9 expression was detected in pure human GBM xenografts. Immunocytochemistry studies showed TLR9 expression in some macrophages/microglial cells. The expression of TLR9 within human GBM strengthens the rationale for the utilization of CpG-ODN in this disease.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Toll-Like Receptor 9/biosynthesis , Toll-Like Receptor 9/genetics , Adult , Aged , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Female , Fluorescent Antibody Technique , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunohistochemistry , Karnofsky Performance Status , Lymphocytes/pathology , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Transplantation , Neurosurgical Procedures , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival
9.
J Neurooncol ; 83(2): 173-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17219056

ABSTRACT

MGMT promoter methylation, which has been correlated with the response to alkylating agents, was investigated in a retrospective series of 219 glioblastomas (GBMs) treated with various modalities. MGMT methylation had no impact on survival for the whole group, but showed a significant advantage (17.1 months vs. 13.1) for patients treated with RT+ adjuvant chemotherapy (relative risk of death (RR) = 0.53; P = 0.041), particularly when patients received CT during the course of RT (MS = 19.9 months vs. 12.5 months; RR = 0.227, P = 0.001). This suggests that the prognostic impact of MGMT methylation is dependent on therapeutic modalities and schedules. MGMT methylation was not correlated with the main molecular alterations, such as 10q loss and p53 expression.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Glioblastoma/metabolism , Tumor Suppressor Proteins/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Chemotherapy, Adjuvant , Combined Modality Therapy , Drug Administration Schedule , Female , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Humans , In Vitro Techniques , Methylation , Middle Aged , Prognosis , Retrospective Studies , Statistics, Nonparametric , Survival Analysis , Tumor Suppressor Protein p53/metabolism
10.
Cancer Genet Cytogenet ; 172(1): 33-7, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17175377

ABSTRACT

The A/G61 polymorphism located in the 5'UTR of the EGF gene has been found to be both a risk factor and a prognostic factor in glioblastoma (GBM), but the functional consequences have not been investigated. Here we show, in vitro, that this polymorphism is functional, in that the G allele promoter is 40% more active than the A variant (P < 0.001). However, analysis of a large series of 209 GBM patients and 214 control subjects did not confirm that A/G61 polymorphism is a significant risk factor for GBM, despite a trend for higher GG frequency in these patients. Furthermore, A/G61 polymorphism was not a prognostic factor for survival in GBM patients, although it does appear to affect progression-free survival.


Subject(s)
Brain Neoplasms/genetics , Epidermal Growth Factor/genetics , Glioblastoma/genetics , Polymorphism, Genetic/genetics , 5' Untranslated Regions/genetics , Adult , Aged , Aged, 80 and over , Animals , CHO Cells , Case-Control Studies , Cricetinae , DNA, Neoplasm/genetics , Epidermal Growth Factor/metabolism , Genetic Predisposition to Disease , Genotype , Humans , Middle Aged , Prognosis , Risk Factors , Survival Rate , Transfection
11.
Ann Neurol ; 60(6): 740-3, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17192931

ABSTRACT

The methylation status of the O6-methylguanine-methyltransferase promoter (MGMTP) was evaluated in 68 low-grade gliomas treated by neoadjuvant temozolomide. Methylated MGMTP was detected in 63 of 68 (92.6 %) patients and was a favorable predictor of progression-free survival as compared with unmethylated MGMTP tumors (p < 0.0001). Assessment of MGMTP status could help identifying low-grade gliomas patients more likely to respond to chemotherapy or to benefit from MGMT depletion strategies.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/drug therapy , DNA Methylation/drug effects , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Promoter Regions, Genetic/drug effects , Tumor Suppressor Protein p14ARF/genetics , Adult , Aged , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , DNA Modification Methylases , DNA Mutational Analysis , DNA Repair Enzymes , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Disease Progression , Female , Genetic Markers/drug effects , Genetic Markers/genetics , Glioma/diagnosis , Glioma/genetics , Humans , Male , Middle Aged , Neoadjuvant Therapy , Predictive Value of Tests , Prognosis , Promoter Regions, Genetic/genetics , Survival Rate , Temozolomide , Tumor Suppressor Proteins
12.
Mol Cancer Ther ; 5(9): 2182-92, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16985051

ABSTRACT

Gliomas are highly lethal neoplasms that cannot be cured by currently available therapies. Temozolomide is a recently introduced alkylating agent that has yielded a significant benefit in the treatment of high-grade gliomas. However, either de novo or acquired chemoresistance occurs frequently and has been attributed to increased levels of O6-methylguanine-DNA methyltransferase or to the loss of mismatch repair capacity. However, very few gliomas overexpress O6-methylguanine-DNA methyltransferase or are mismatch repair-deficient, suggesting that other mechanisms may be involved in the resistance to temozolomide. The purpose of the present study was to generate temozolomide-resistant variants from a human glioma cell line (SNB-19) and to use large-scale genomic and transcriptional analyses to study the molecular basis of acquired temozolomide resistance. Two independently obtained temozolomide-resistant variants exhibited no cross-resistance to other alkylating agents [1,3-bis(2-chloroethyl)-1-nitrosourea and carboplatin] and shared genetic alterations, such as loss of a 2p region and loss of amplification of chromosome 4 and 16q regions. The karyotypic alterations were compatible with clonal selection of preexistent resistant cells in the parental SNB-19 cell line. Microarray analysis showed that 78 out of 17,000 genes were differentially expressed between parental cells and both temozolomide-resistant variants. None are implicated in known resistance mechanisms, such as DNA repair, whereas interestingly, several genes involved in differentiation were down-regulated. The data suggest that the acquisition of resistance to temozolomide in this model resulted from the selection of less differentiated preexistent resistant cells in the parental tumor.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Glioma/genetics , Antineoplastic Agents, Alkylating/pharmacokinetics , Cell Line, Tumor , Dacarbazine/pharmacokinetics , Dacarbazine/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression , Glioma/metabolism , Humans , In Situ Hybridization, Fluorescence/methods , Karyotyping/methods , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , O(6)-Methylguanine-DNA Methyltransferase/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Temozolomide
13.
Cancer ; 107(8): 1891-7, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16986124

ABSTRACT

BACKGROUND: There is a need to improve the current, controversial, and poorly reproducible classification of anaplastic gliomas, which represent a highly heterogeneous entity in terms of survival. METHODS: The impact of the most common genetic alterations on survival was investigated based on 156 anaplastic gliomas: Among the patients who were included, the gender ratio was 1.32, the median age was 45.5 years (range, 20-83 years), and the median Karnofsky performance status was 70 (range, 40-100). Genetic analysis included a search for loss of heterozygosity (LOH) on chromosomes 1p and 19q; amplification of chromosomes 9p and 10q and of the epidermal growth factor receptor (EGFR), cyclin-dependent kinase 4 (CDK4) and mouse double-minute (MDM2) genes; and p53 expression. RESULTS: The median survival was 33.5 months, and the median progression-free survival was 15.8 months. In a univariate analysis, LOH on 1p and 19q was correlated with longer survival, whereas p53 expression, LOH on 9p, LOH on 10q, amplified EGFR, and deleted CDKN2A were correlated with shorter survival. LOH on 1p and 19q were associated with oligodendrogliomas, LOH on 10q was related to EGFR amplification, and LOH on 1p and 19q was mutually exclusive with EGFR amplification and LOH on 10q. In a multivariate analysis, the significant prognostic factors were age, histology, LOH on 1p and 19q, and P16/CDKN2A deletion. Recursive partitioning analysis (RPA) divided the whole group hierarchically into 3 distinct prognostic subgroups: Group A with 1p19q codeletion (median survival, 98 months), Group B with EGFR amplification (median survival, 17 months), and Group CC (median survival, 31 months), providing a basis for a genetically based prognostic subclassification for patients with Grade III gliomas. CONCLUSIONS: The search for 1p19q codeletion and EGFR receptor amplification provides a simple, clinically relevant prognostic subclassification of grade III gliomas.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Adult , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 10 , Chromosomes, Human, Pair 9 , Disease-Free Survival , ErbB Receptors/genetics , Female , Glioma/mortality , Humans , Loss of Heterozygosity , Male , Middle Aged , Molecular Diagnostic Techniques , Prognosis , Survival Analysis , Tumor Suppressor Protein p53/metabolism
14.
Cancer ; 106(10): 2218-23, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16568472

ABSTRACT

BACKGROUND: In contrast to oligodendrogliomas, molecular predictors of prognosis have not been consistently found in glioblastomas. However, genetic studies show that glioblastomas consist of several genetic subtypes and raise the possibility that molecular alterations could be predictive of survival. METHODS: A search for loss of heterozygosity (LOH) on chromosome 1p, 9p, 10q, 19q, EGFR (epidermal growth factor receptor), CDK4, and MDM2 (mouse double minute) amplifications, CDKN2A (INK4A/ARF) homozygous deletions, p53 expression, was performed in a series of 220 primary glioblastomas. The molecular alterations were then correlated with each other to identify distinct molecular pathways and with clinical parameters and the course of the disease to identify prognostic markers. RESULTS: Nonrandom associations were found between EGFR amplification and LOH10q, LOH9p, and INK4A/ARF deletion, LOH1p and LOH19q, and MDM2 and CDK4 amplification, whereas mutual exclusions were found between p53 expression and EGFR amplification, LOH 9p/INK4A/ARF homozygous deletion, and MDM2 and CDK4 amplification. Age (P = 4.10(-5)) and performance status (P = .003) were the main predictors of outcome. In contrast, molecular markers were of limited impact: MDM2 amplification correlated with poor outcome on both univariate and multivariate analysis (P = .01) and EGFR amplification with good prognosis on multivariate analysis (P = .02). CONCLUSION: Despite their limited prognostic impact, the genetic markers investigated here outline distinct molecular pathways involved in glioblastoma tumorigenesis and warrant broader molecular screening.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/mortality , Genes, erbB-1/genetics , Glioblastoma/genetics , Glioblastoma/mortality , Proto-Oncogene Proteins c-mdm2/genetics , Adolescent , Adult , Base Sequence , Brain Neoplasms/pathology , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Genetic Markers , Glioblastoma/pathology , Humans , Male , Middle Aged , Molecular Sequence Data , Multivariate Analysis , Neoplasm Staging , Polymerase Chain Reaction , Probability , Prognosis , Proportional Hazards Models , Registries , Retrospective Studies , Risk Assessment , Sensitivity and Specificity , Survival Analysis
15.
Ann Neurol ; 58(2): 322-6, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16049942

ABSTRACT

Search for loss of heterozygosity on chromosomes 1p, 9p, 10q, and 19q, epidermal growth factor receptor (EGFR) gene amplification, and p53 expression was performed in a series of 131 low-grade gliomas. The profile of molecular changes, clinical findings, and histology were subsequently correlated with the course of the disease, mainly progression-free survival. When these parameters were considered as candidate variables in a multivariate analysis, only loss of heterozygosity on chromosome 1p was associated with increased progression-free survival (hazard ratio, 0.521), indicating a major favorable prognostic role of this genetic alteration in low-grade gliomas.


Subject(s)
Brain Neoplasms/genetics , Chromosome Deletion , Chromosomes, Human, Pair 1 , Glioma/genetics , Loss of Heterozygosity , Adolescent , Adult , Aged , Brain Neoplasms/pathology , Chromosomes, Human, Pair 19 , Female , Glioma/pathology , Humans , Immunohistochemistry/methods , Male , Middle Aged , Molecular Biology/methods
16.
Arterioscler Thromb Vasc Biol ; 24(12): 2358-64, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15486308

ABSTRACT

OBJECTIVE: The human scavenger receptor class B type I (Cla-1) plays a key role in cellular cholesterol movement in facilitating transport of cholesterol between cells and lipoproteins. Indirect evidence has suggested that Cla-1 gene expression is under the feedback control of cellular cholesterol content. To define the molecular mechanisms underlying such putative regulation, we evaluated whether Cla-1 is a target gene of the sterol regulatory element binding protein (SREBP) transcription factor family. METHODS AND RESULTS: Transient transfections demonstrated that SREBP factors induce Cla-1 promoter activity and that SREBP-2 is a more potent inducer than the SREBP-1a isoform. The 5'-deletion analysis of 3 kb of the 5'-flanking sequence of the Cla-1 gene, combined with site-directed mutagenesis and electrophoretic mobility shift assay, allowed identification of a unique sterol responsive element. SREBP-mediated Cla-1 regulation was confirmed in stably transfected human embryonic kidney 293 cells expressing the active form of SREBP-2 at incremental levels. In these cell lines, Cla-1 mRNA and protein levels were increased in direct proportion to the level of SREBP-2 expression. CONCLUSIONS: These findings provide evidence that SREBP-2, a key regulator of cellular cholesterol uptake through modulation of the expression of the low-density lipoprotein receptor gene, may influence cellular cholesterol homeostasis via regulation of Cla-1 gene expression.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation/physiology , Receptors, Immunologic/genetics , Transcription Factors/physiology , Binding Sites/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Kidney/chemistry , Kidney/cytology , Kidney/embryology , Kidney/metabolism , Mutagenesis, Site-Directed/genetics , Promoter Regions, Genetic/genetics , Receptors, Immunologic/metabolism , Receptors, Scavenger , Recombinant Proteins/genetics , Scavenger Receptors, Class B , Sterol Regulatory Element Binding Protein 1 , Sterol Regulatory Element Binding Protein 2 , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection/methods
17.
Arterioscler Thromb Vasc Biol ; 24(11): 2192-5, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15358600

ABSTRACT

OBJECTIVE: A common polymorphism in the promoter of the apolipoprotein B (apoB) gene, a C to T change at position -516, increases the transcription rate of apoB, resulting in elevated circulating levels of low-density lipoprotein (LDL) cholesterol. METHODS AND RESULTS: We tested the hypothesis that carriers of the -516T allele, who may display consistent elevation in plasma cholesterol over their lifetime, may present more extensive atherosclerotic disease than noncarriers. Genotyping of the apoB 516 C/T promoter polymorphism was performed in 326 subjects at low cardiovascular risk. Homozygotes for allele T displayed higher plasma levels of apoB and LDL than did heterozygotes. Furthermore, both homozygotes and heterozygotes for allele T exhibited higher plasma levels of apoB and LDL than did homozygotes for allele C (P<0.0001). In addition, homozygotes for allele T displayed higher carotid intima-media thickness (IMT) than subjects who were heterozygous. Moreover, both groups had higher carotid IMT than subjects of genotype -516C/C (P<0.001). Only age, high-density lipoprotein, and the presence of allele T were identified as independent predictors of the presence of carotid plaque. No association existed between the polymorphism and plasma concentrations of triglycerides, high-density lipoprotein, or apoAI. CONCLUSIONS: Our data indicate that a C to T change at position -516 of the apoB gene is independently associated with the presence of carotid atherosclerotic disease. Identification of the -516C/T polymorphism may therefore contribute to the estimation of overall cardiovascular risk.


Subject(s)
Apolipoproteins B/genetics , Carotid Artery Diseases/blood , Carotid Artery Diseases/genetics , Cholesterol/blood , Cytosine/metabolism , Polymorphism, Genetic/genetics , Promoter Regions, Genetic/genetics , Thymine/metabolism , Apolipoproteins/blood , Female , Genetic Predisposition to Disease/genetics , Genotype , Humans , Lipids/blood , Male , Middle Aged , Predictive Value of Tests , Tunica Intima/pathology , Tunica Media/pathology
19.
Curr Opin Oncol ; 16(6): 607-13, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15627025

ABSTRACT

PURPOSE OF REVIEW: Despite optimal clinical treatment, the prognosis for gliomas remains poor, and little progress has been observed during the last few years. Meanwhile, understanding of glioma oncogenesis has improved greatly. This review focuses on recent advances in molecular biology of glial tumors, with particular emphasis on lineage markers, genetic mechanisms underlying tumor progression, new diagnostic and prognostic markers, and potential therapeutic targets. RECENT FINDINGS: The question of the cell of origin, illustrated by the evidence of tumor-derived multipotent progenitors, by the animal models of gliomas, and by lineage markers such as Olig1/2 markers, remains unsolved. Genotype/phenotype correlation studies have identified early and late genetic alterations related either to astrocytic or oligodendroglial phenotype. They complement the existing World Health Organization morphologic classification and provide additional prognostic markers such as 1p/19q deletion in oligodendrogliomas. Most of these genetic alterations result in the disruption of three main cellular systems: RB1, P53, and tyrosine kinase receptor pathways. New gene alterations have also been identified in glioma, promoting mitotic signal transduction, cell cycle regulation, apoptosis, angiogenesis, or invasion. Gene and protein profiling has been correlated with outcome. SUMMARY: Management of gliomas, especially oligodendrogliomas with 1p19q deletion, benefits from advances in molecular genetics. A better understanding of the molecular pathogenesis and cellular lineage of gliomas will improve tumor classification and define more reliable prognostic markers. There is a hope that it will also lead to novel targets for therapy.


Subject(s)
Central Nervous System Neoplasms/genetics , Glioma/genetics , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/therapy , Epidermal Growth Factor/antagonists & inhibitors , Gefitinib , Glioma/diagnosis , Glioma/therapy , Humans , Neovascularization, Pathologic/genetics , Quinazolines/therapeutic use
20.
Arterioscler Thromb Vasc Biol ; 23(9): 1633-9, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12842837

ABSTRACT

OBJECTIVE: The apolipoprotein(a) [apo(a)] gene locus is the major determinant of the circulating concentration of the atherothrombogenic lipoprotein Lp(a). In vitro analysis of the intergenic region between the apo(a) and plasminogen genes revealed the presence of a putative apo(a) transcription control region (ACR) approximately 20 kb upstream of the apo(a) gene that significantly increases the minimal promoter activity of the human apo(a) gene. METHODS AND RESULTS: To examine the function of the ACR in its natural genomic context, we used the Cre-loxP recombination system to generate 2 nearly identical apo(a)-yeast artificial chromosome transgenic mouse lines that possess a single integration site for the human apo(a) transgene in the mouse genome but differ by the presence or absence of the ACR enhancer. Analysis of the 2 groups of animals revealed that the deletion of the ACR was associated with 30% reduction in plasma and mRNA apo(a) levels. Apo(a)-yeast artificial chromosome transgenic mice with and without the ACR sequence were similar in all other aspects of apo(a) regulation, including liver-specific apo(a) expression and alteration in expression levels in response to sexual maturation and a high-fat diet. CONCLUSIONS: This study provides the first experimental in vivo evidence for a functional role of the ACR enhancer in determining levels of apo(a) expression.


Subject(s)
5' Untranslated Regions/physiology , Apolipoproteins A/genetics , Chromosomes, Artificial, Yeast/genetics , Enhancer Elements, Genetic/physiology , Gene Expression Regulation/physiology , Promoter Regions, Genetic/genetics , Transcription, Genetic/physiology , Transgenes , 5' Untranslated Regions/genetics , Animals , Blastocyst/chemistry , Blastocyst/metabolism , Chimera , Diet, Atherogenic , Dietary Fats/pharmacology , Enhancer Elements, Genetic/drug effects , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Transfer Techniques , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , Organ Specificity/genetics , Transcription, Genetic/genetics , Transgenes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...