Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Oncol ; 9: 103, 2019.
Article in English | MEDLINE | ID: mdl-30931252

ABSTRACT

The heterogeneous and invasive nature of pediatric gliomas poses significant treatment challenges, highlighting the importance of identifying novel chemotherapeutic targets. Recently, recurrent Fibroblast growth factor receptor 1 (FGFR1) mutations in pediatric gliomas have been reported. Here, we explored the clinical relevance of FGFR1 expression, cell migration in low and high grade pediatric gliomas and the role of FGFR1 in cell migration/invasion as a potential chemotherapeutic target. A high density tissue microarray (TMA) was used to investigate associations between FGFR1 and activated phosphorylated FGFR1 (pFGFR1) expression and various clinicopathologic parameters. Expression of FGFR1 and pFGFR1 were measured by immunofluorescence and by immunohistochemistry (IHC) in 3D spheroids in five rare patient-derived pediatric low-grade glioma (pLGG) and two established high-grade glioma (pHGG) cell lines. Two-dimensional (2D) and three-dimensional (3D) migration assays were performed for migration and inhibitor studies with three FGFR1 inhibitors. High FGFR1 expression was associated with age, malignancy, tumor location and tumor grade among astrocytomas. Membranous pFGFR1 was associated with malignancy and tumor grade. All glioma cell lines exhibited varying levels of FGFR1 and pFGFR1 expression and migratory phenotypes. There were significant anti-migratory effects on the pHGG cell lines with inhibitor treatment and anti-migratory or pro-migratory responses to FGFR1 inhibition in the pLGGs. Our findings support further research to target FGFR1 signaling in pediatric gliomas.

2.
Cancer Lett ; 346(1): 34-44, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24333734

ABSTRACT

Promoter hypermethylation and transcriptional silencing is a common epigenetic mechanism of gene inactivation in cancer. To identify targets of epigenetic silencing in paediatric intracranial ependymoma, we used a pharmacological unmasking approach through treatment of 3 ependymoma short-term cell cultures with the demethylating agent 5-Aza-2'-deoxycytidine followed by global expression microarray analysis. We identified 55 candidate epigenetically silenced genes, which are involved in the regulation of apoptosis, Wnt signalling, p53 and cell differentiation. The methylation status of 26 of these genes was further determined by combined bisulfite restriction analysis (COBRA) and genomic sequencing in a cohort of 40 ependymoma samples. The most frequently methylated genes were BEX1 (27/40 cases), BAI2 (20/40), CCND2 (18/40), and CDKN2A (14/40). A high correlation between promoter hypermethylation and decreased gene expression levels was established by real-time quantitative PCR, suggesting the involvement of these genes in ependymoma tumourigenesis. Furthermore, ectopic expression of brain-expressed X-linked 1 (BEX1) in paediatric ependymoma short-term cell cultures significantly suppressed cell proliferation and colony formation. These data suggest that promoter hypermethylation contributes to silencing of target genes in paediatric intracranial ependymoma. Epigenetic inactivation of BEX1 supports its role as a candidate tumour suppressor gene in intracranial ependymoma, and a potential target for novel therapies for ependymoma in children.


Subject(s)
Brain Neoplasms/genetics , Ependymoma/genetics , Epigenesis, Genetic , Nerve Tissue Proteins/genetics , Cells, Cultured , Child , DNA Methylation , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Genome-Wide Association Study , Humans , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Transfection
3.
Bioorg Med Chem ; 21(7): 1918-24, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23419325

ABSTRACT

A series of related 2-arylindoles have been evaluated for their anticancer activity against a range of glioblastoma cell lines using a number of different cell-based assays to determine cell viability after treatment with the compounds. The best indoles, which showed comparable activity to cisplatin against a U87MG cell line in the MTS assay, were taken forward and initial studies suggest that their mechanism of action is consistent with the generation of reactive oxygen species followed by autophagic cell death. Furthermore, activity was also observed in glioblastoma short-term cell cultures for the best lead compound and in some cases gave low micromolar IC50s.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Glioblastoma/drug therapy , Indoles/chemistry , Indoles/pharmacology , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Glioblastoma/metabolism , Humans , Inhibitory Concentration 50 , Reactive Oxygen Species/metabolism
4.
J Neurooncol ; 98(3): 305-18, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20052518

ABSTRACT

Little is known about the cytogenetic and molecular genetic events that lead to the formation of paediatric astrocytoma. We have analysed 57 paediatric astrocytoma (WHO grades I-IV) using comparative genomic hybridisation in order to identify common regions of abnormality. Large regions of copy number alterations were infrequent with 71% of tumours demonstrating no genomic imbalance. Furthermore, the most frequent aberrations (including gain of 6q, 2q, and 7q, and loss of 16 and 12q) occurred in only a subset of cases. High-copy number amplification was seen in five tumours at 12 different regions. The presence of copy number alterations was significantly associated with increasing grade of malignancy, and gain of 12q and the presence of high-copy number amplification were associated with a poor outcome in patients with malignant astrocytoma (P = 0.0039 and 0.0085, respectively). FISH analysis confirmed loss of 1p36 identified by CGH. There was no evidence of amplification of EGFR, CDK4, MET, CDK6, c-myc, or MDM2.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Comparative Genomic Hybridization/methods , In Situ Hybridization, Fluorescence/methods , Adolescent , Analysis of Variance , Astrocytoma/classification , Astrocytoma/mortality , Brain Neoplasms/classification , Brain Neoplasms/mortality , Child , Child, Preschool , Chromosome Aberrations , Chromosome Disorders/genetics , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 7/genetics , Chromosomes, Human, Pair 8/genetics , Female , Humans , Infant , Male , Pediatrics , Survival Analysis , Young Adult
5.
Genes Chromosomes Cancer ; 47(11): 1005-22, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18663750

ABSTRACT

Loss of chromosome 22 and gain of 1q are the most frequent genomic aberrations in ependymomas, indicating that genes mapping to these regions are critical in their pathogenesis. Using real-time quantitative PCR, we measured relative copy numbers of 10 genes mapping to 22q12.3-q13.33 and 10 genes at 1q21-32 in a series of 47 pediatric intracranial ependymomas. Loss of one or more of the genes on 22 was detected in 81% of cases, with RAC2 and C22ORF2 at 22q12-q13.1 being deleted most frequently in 38% and 32% of ependymoma samples, respectively. Combined analysis of quantitative-PCR with methylation-specific PCR and bisulphite sequencing revealed a high rate (>60% ependymoma) of transcriptional inactivation of C22ORF2, indicating its potential importance in the development of pediatric ependymomas. Increase of relative copy numbers of at least one gene on 1q were detected in 61% of cases, with TPR at 1q25 displaying relative copy number gains in 38% of cases. Patient age was identified as a significant adverse prognostic factor, as a significantly shorter overall survival time (P = 0.0056) was observed in patients <2 years of age compared with patients who were >2 years of age. Loss of RAC2 at 22q13 or amplification of TPR at 1q25 was significantly associated with shorter overall survival in these younger patients (P = 0.0492 and P = < 0.0001, respectively). This study identifies candidate target genes within 1q and 22q that are potentially important in the pathogenesis of intracranial pediatric ependymomas.


Subject(s)
Brain Neoplasms/genetics , Carrier Proteins/genetics , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 22/genetics , Ependymoma/genetics , Nuclear Pore Complex Proteins/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Adolescent , Base Sequence , Biomarkers, Tumor/genetics , Child , Child, Preschool , DNA, Neoplasm/genetics , Female , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction/methods
6.
Ann N Y Acad Sci ; 1031: 169-83, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15753143

ABSTRACT

Colorectal cancer (CRC) is responsible for the second highest associated mortality in Western Europe and the United States. Approximately 95% of CRC is sporadic and believed to involve environmental agents and chronic inflammation as causal elements. Several recent studies have suggested a link with diet, in particular, red meat, dietary fats, and low consumption of vegetables. Lipid peroxidation and arachidonic acid metabolism have specifically been implicated in genotoxicity, tumor initiation, and promotion. We have examined the global gene expression profiles (Affymetrix; HU133A) of differentiated vs. undifferentiated colonocytes (CRL-1807), with and without vitamin E supplementation, while undergoing a lipid peroxidative stress. Malondialdehyde and hydroxynonenal, generated by heating a mixture of linoleic and linolenic acid, caused DNA adduct formation identified by immunofluoresence. We also observed a decreased ability for vitamin E to upregulate detoxifying enzymes against free-radical peroxidation, with the exception of mitochondrial superoxide dismutase in undifferentiated cells. However, there was an increased ability in undifferentiated, rather than in differentiated, colonic cells to detect DNA damage, initiate cytostasis, and then effect subsequent DNA repair and apoptosis, in the presence of vitamin E. The expression profile implies less genotoxic stress is experienced in vitamin E-supplemented colonocytes, particularly undifferentiated cells, and points to a mechanism by which dietary supplementation may prevent genotoxic damage and subsequent carcinogenic events in the colon, by both antioxidant and non-antioxidant-related mechanisms.


Subject(s)
Colonic Neoplasms/etiology , Colonic Neoplasms/genetics , Diet , Gene Expression/drug effects , Vitamin E/pharmacology , Anticarcinogenic Agents/pharmacology , Antioxidants/pharmacology , Arachidonic Acid/metabolism , DNA Adducts/analysis , Dietary Supplements , Gene Expression Profiling , Humans , Lipid Peroxidation , Oligonucleotide Array Sequence Analysis , Oxidative Stress/drug effects , RNA, Messenger/analysis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...