Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Clin Epigenetics ; 11(1): 144, 2019 Oct 21.
Article in English | MEDLINE | ID: mdl-31639040

ABSTRACT

After publication of the original article [1], authors have requested to add a 'J' as middle name for Richard Gilbertson. Hence, full name should be Richard J Gilbertson.

2.
Clin Epigenetics ; 11(1): 117, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31409384

ABSTRACT

BACKGROUND: Histological grading of choroid plexus tumors (CPTs) remains the best prognostic tool to distinguish between aggressive choroid plexus carcinoma (CPC) and the more benign choroid plexus papilloma (CPP) or atypical choroid plexus papilloma (aCPP); however, these distinctions can be challenging. Standard treatment of CPC is very aggressive and often leads to severe damage to the young child's brain. Therefore, it is crucial to distinguish between CPC and less aggressive entities (CPP or aCPP) to avoid unnecessary exposure of the young patient to neurotoxic therapy. To better stratify CPTs, we utilized DNA methylation (DNAm) to identify prognostic epigenetic biomarkers for CPCs. METHODS: We obtained DNA methylation profiles of 34 CPTs using the HumanMethylation450 BeadChip from Illumina, and the data was analyzed using the Illumina Genome Studio analysis software. Validation of differentially methylated CpG sites chosen as biomarkers was performed using pyrosequencing analysis on additional 22 CPTs. Sensitivity testing of the CPC DNAm signature was performed on a replication cohort of 61 CPT tumors obtained from Neuropathology, University Hospital Münster, Germany. RESULTS: Generated genome-wide DNAm profiles of CPTs showed significant differences in DNAm between CPCs and the CPPs or aCPPs. The prediction of clinical outcome could be improved by combining the DNAm profile with the mutational status of TP53. CPCs with homozygous TP53 mutations clustered as a group separate from those carrying a heterozygous TP53 mutation or CPCs with wild type TP53 (TP53-wt) and showed the worst survival outcome. Specific DNAm signatures for CPCs revealed AK1, PER2, and PLSCR4 as potential biomarkers for CPC that can be used to improve molecular stratification for diagnosis and treatment. CONCLUSIONS: We demonstrate that combining specific DNAm signature for CPCs with histological approaches better differentiate aggressive tumors from those that are not life threatening. These findings have important implications for future prognostic risk prediction in clinical disease management.


Subject(s)
Adenylate Kinase/genetics , Choroid Plexus Neoplasms/diagnosis , DNA Methylation , Epigenomics/methods , Period Circadian Proteins/genetics , Phospholipid Transfer Proteins/genetics , Biomarkers, Tumor/genetics , Carcinoma/diagnosis , Carcinoma/genetics , Carcinoma/mortality , Choroid Plexus Neoplasms/genetics , Choroid Plexus Neoplasms/mortality , CpG Islands , Diagnosis, Differential , Epigenesis, Genetic , Humans , Mutation , Papilloma, Choroid Plexus/diagnosis , Papilloma, Choroid Plexus/genetics , Papilloma, Choroid Plexus/mortality , Prognosis , Survival Analysis , Tumor Suppressor Protein p53/genetics
3.
Clin Cancer Res ; 21(1): 184-92, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25336695

ABSTRACT

PURPOSE: To investigate molecular alterations in choroid plexus tumors (CPT) using a genome-wide high-throughput approach to identify diagnostic and prognostic signatures that will refine tumor stratification and guide therapeutic options. EXPERIMENTAL DESIGN: One hundred CPTs were obtained from a multi-institutional tissue and clinical database. Copy-number (CN), DNA methylation, and gene expression signatures were assessed for 74, 36, and 40 samples, respectively. Molecular subgroups were correlated with clinical parameters and outcomes. RESULTS: Unique molecular signatures distinguished choroid plexus carcinomas (CPC) from choroid plexus papillomas (CPP) and atypical choroid plexus papillomas (aCPP); however, no significantly distinct molecular alterations between CPPs and aCPPs were observed. Allele-specific CN analysis of CPCs revealed two novel subgroups according to DNA content: hypodiploid and hyperdiploid CPCs. Hyperdiploid CPCs exhibited recurrent acquired uniparental disomy events. Somatic mutations in TP53 were observed in 60% of CPCs. Investigating the number of mutated copies of p53 per sample revealed a high-risk group of patients with CPC carrying two copies of mutant p53, who exhibited poor 5-year event-free (EFS) and overall survival (OS) compared with patients with CPC carrying one copy of mutant p53 (OS: 14.3%, 95% confidence interval, 0.71%-46.5% vs. 66.7%, 28.2%-87.8%, respectively, P = 0.04; EFS: 0% vs. 44.4%, 13.6%-71.9%, respectively, P = 0.03). CPPs and aCPPs exhibited favorable survival. DISCUSSION: Our data demonstrate that differences in CN, gene expression, and DNA methylation signatures distinguish CPCs from CPPs and aCPPs; however, molecular similarities among the papillomas suggest that these two histologic subgroups are indeed a single molecular entity. A greater number of copies of mutated TP53 were significantly associated to increased tumor aggressiveness and a worse survival outcome in CPCs. Collectively, these findings will facilitate stratified approaches to the clinical management of CPTs.


Subject(s)
Choroid Plexus Neoplasms/genetics , DNA Methylation/genetics , Neoplasm Proteins/biosynthesis , Prognosis , Adolescent , Child , Child, Preschool , Choroid Plexus Neoplasms/classification , Choroid Plexus Neoplasms/pathology , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Infant , Male , Mutation , Neoplasm Staging , Tumor Suppressor Protein p53/genetics
4.
J Clin Oncol ; 28(12): 1995-2001, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20308654

ABSTRACT

PURPOSE Choroid plexus carcinomas are pediatric tumors with poor survival rates and a strong, but poorly understood, association with Li-Fraumeni syndrome (LFS). Currently, with lack of biologic predictors, most children are treated with aggressive chemoradiation protocols. PATIENTS AND METHODS We established a multi-institutional tissue and clinical database, which enabled the analysis of specific alterations of the TP53 tumor suppressor and its modifiers in choroid plexus tumors (CPTs). We conducted high-resolution copy-number analysis to correlate these genetic parameters with family history and outcome. Results We studied 64 patients with CPTs. All individuals with germline TP53 mutations fulfilled LFS criteria, whereas all patients not meeting these criteria harbored wild-type TP53 (P < .001). TP53 mutations were found in 50% of choroid plexus carcinomas (CPCs). Additionally, two sequence variants known to confer TP53 dysfunction, TP53 codon72 and MDM2 SNP309, coexisted in the majority of TP53 wild-type CPCs (92%) and not in TP53 mutated CPC (P = .04), which suggests a complementary mechanism of TP53 dysfunction in the absence of a TP53 mutation. High-resolution single nucleotide polymorphism (SNP) array analysis revealed extremely high total structural variation (TSV) in TP53-mutated CPC tumor genomes compared with TP53 wild-type tumors and choroid plexus papillomas (CPPs; P = .006 and .004, respectively). Moreover, high TSV was associated with significant risk of progression (P < .001). Five-year survival rates for patients with TP53-immunopositive and -immunonegative CPCs were 0% and 82 (+/- 9%), respectively (P < .001). Furthermore, 14 of 16 patients with TP53 wild-type CPCs are alive without having received radiation therapy. CONCLUSION Patients with CPC who have low tumor TSV and absence of TP53 dysfunction have a favorable prognosis and can be successfully treated without radiation therapy.


Subject(s)
Carcinoma/genetics , Choroid Plexus Neoplasms/genetics , Germ-Line Mutation , Papilloma, Choroid Plexus/genetics , Polymorphism, Single Nucleotide , Tumor Suppressor Protein p53/genetics , Carcinoma/chemistry , Carcinoma/mortality , Carcinoma/therapy , Chi-Square Distribution , Child , Child, Preschool , Choroid Plexus Neoplasms/chemistry , Choroid Plexus Neoplasms/mortality , Choroid Plexus Neoplasms/therapy , Databases as Topic , Disease-Free Survival , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Oligonucleotide Array Sequence Analysis , Ontario/epidemiology , Papilloma, Choroid Plexus/chemistry , Papilloma, Choroid Plexus/mortality , Papilloma, Choroid Plexus/therapy , Phenotype , Time Factors , Treatment Outcome , Tumor Suppressor Protein p53/analysis , United States/epidemiology
5.
Pediatr Dev Pathol ; 13(3): 169-77, 2010.
Article in English | MEDLINE | ID: mdl-19586133

ABSTRACT

Pediatric undifferentiated soft tissue sarcomas (USTSs) are tumors composed of primitive mesenchymal cells. As such, they form an attractive model for studying the early events in sarcoma development. In an effort to better understand the critical molecular aberrations leading to sarcoma development, gene expression array analysis and post-array validation techniques were applied to several USTSs; the results were consistent with upregulation of the excitatory components of the insulin-like growth factor (IGF) pathway. Particularly high expression of the insulin-like growth factor 2 (IGF2) ligand was seen and confirmed by real-time reverse transcriptase-polymerase chain reaction. Immunohistochemistry performed using antibodies against IGF2 revealed overexpression of the IGF2 protein in 19 of 21 (90%) USTSs and revealed 2 distinct staining patterns, 1 in which there was diffuse cytoplasmic staining (16/19) and 1 in which there was punctate perinuclear positivity (3/19). Using ultrastructural immunogold localization of IGF2, it was determined that IGF2 was primarily localized to Golgi-derived vesicles and multivesicular bodies in tumor cells showing the punctate pattern, and to both the cytoplasm and plasma membrane of tumor cells showing the diffuse pattern. The results suggest that upregulation of the IGF pathway in pediatric USTSs is a critical early event in the development of sarcomas. Furthermore, findings from the immunocytochemical and immunogold analyses confirm the presence of 2 different cytoplasmic trafficking patterns and storage motifs of IGF2 within this type of tumor. Given that in one subcellular pattern the IGF2 protein does not appear to reach the membrane, these findings may have functional significance.


Subject(s)
Cytoplasm/metabolism , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor II/metabolism , Sarcoma/metabolism , Soft Tissue Neoplasms/metabolism , Biomarkers/metabolism , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Child , Cytoplasm/ultrastructure , Gene Expression Profiling , Humans , Immunohistochemistry/methods , Insulin-Like Growth Factor II/genetics , Microscopy, Electron, Transmission , Oligonucleotide Array Sequence Analysis/methods , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma/genetics , Sarcoma/ultrastructure , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/ultrastructure , Tissue Array Analysis , Up-Regulation
6.
J Clin Invest ; 119(6): 1558-70, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19478459

ABSTRACT

Although most reports describe the protein kinase integrin-linked kinase (ILK) as a proto-oncogene, occasional studies detail opposing functions in the regulation of normal and transformed cell proliferation, differentiation, and apoptosis. Here, we demonstrated that ILK functions as an oncogene in the highly aggressive pediatric sarcoma alveolar rhabdomyosarcoma (ARMS) and as a tumor suppressor in the related embryonal rhabdomyosarcoma (ERMS). These opposing functions hinge on signaling through a noncanonical ILK target, JNK1, to the proto-oncogene c-Jun. RNAi-mediated depletion of ILK induced activation of JNK and its target, c-Jun, resulting in growth of ERMS cells, whereas in ARMS cells, it led to loss of JNK/c-Jun signaling and suppression of growth both in vitro and in vivo. Ectopic expression of the fusion gene characteristic of ARMS (paired box 3-forkhead homolog in rhabdomyosarcoma [PAX3-FKHR]) in ERMS cells was sufficient to convert them to an ARMS signaling phenotype and render ILK activity oncogenic. Furthermore, restoration of JNK1 in ARMS reestablished a tumor-suppressive function for ILK. These findings indicate what we believe to be a novel effector pathway regulated by ILK, provide a mechanism for interconversion of oncogenic and tumor-suppressor functions of a single regulatory protein based on the genetic background of the tumor cells, and suggest a rationale for tailored therapy of rhabdomyosarcoma based on the different activities of ILK.


Subject(s)
Mitogen-Activated Protein Kinase 8/metabolism , Oncogene Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Rhabdomyosarcoma/enzymology , Tumor Suppressor Proteins/metabolism , Animals , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Oncogene Proteins, Fusion/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Mas , RNA, Small Interfering/genetics , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Signal Transduction , Survival Rate , Xenograft Model Antitumor Assays
7.
Proc Natl Acad Sci U S A ; 105(32): 11264-9, 2008 Aug 12.
Article in English | MEDLINE | ID: mdl-18685109

ABSTRACT

DNA copy number variations (CNVs) are a significant and ubiquitous source of inherited human genetic variation. However, the importance of CNVs to cancer susceptibility and tumor progression has not yet been explored. Li-Fraumeni syndrome (LFS) is an autosomal dominantly inherited disorder characterized by a strikingly increased risk of early-onset breast cancer, sarcomas, brain tumors and other neoplasms in individuals harboring germline TP53 mutations. Known genetic determinants of LFS do not fully explain the variable clinical phenotype in affected family members. As part of a wider study of CNVs and cancer, we conducted a genome-wide profile of germline CNVs in LFS families. Here, by examining DNA from a large healthy population and an LFS cohort using high-density oligonucleotide arrays, we show that the number of CNVs per genome is well conserved in the healthy population, but strikingly enriched in these cancer-prone individuals. We found a highly significant increase in CNVs among carriers of germline TP53 mutations with a familial cancer history. Furthermore, we identified a remarkable number of genomic regions in which known cancer-related genes coincide with CNVs, in both LFS families and healthy individuals. Germline CNVs may provide a foundation that enables the more dramatic chromosomal changes characteristic of TP53-related tumors to be established. Our results suggest that screening families predisposed to cancer for CNVs may identify individuals with an abnormally high number of these events.


Subject(s)
Chromosome Aberrations , Genetic Predisposition to Disease , Genome, Human/genetics , Germ-Line Mutation , Li-Fraumeni Syndrome/genetics , Tumor Suppressor Protein p53/genetics , Cohort Studies , DNA/genetics , Female , Humans , Male , Oligonucleotide Array Sequence Analysis/methods
8.
Oncogene ; 22(3): 471-5, 2003 Jan 23.
Article in English | MEDLINE | ID: mdl-12545170

ABSTRACT

Ataxia telangiectasia (AT) is a human hereditary syndrome whose underlying gene product, ataxia telangiectasia mutated (ATM) protein kinase, is involved in multiple intracellular signaling pathways. We demonstrated previously that AT fibroblasts are defective in intracellular Ca(2+) mobilization in response to both stress-inducing and mitogenic stimuli. To extend these findings, normal and AT cells were exposed to serum in the presence of different concentrations of extracellular Ca(2+) ([Ca(2+)](o)), and release of intracellular Ca(2+), activation of calmodulin-dependent protein kinase II and phosphorylation of kinases ERK1 and 2 were monitored. When maintained in high [Ca(2+)](o) (0.42 mM), normal fibroblasts responded to serum introduction more rapidly and efficiently than did AT cells. Unexpectedly, decreasing the [Ca(2+)](o) in the medium had a diametrically opposite effect. Under low [Ca(2+)](o) (0.0022 mM) conditions, normal cells were slow and inefficient in their responses, whereas AT cells showed a substantial improvement in all three end points. These findings demonstrate that loss of ATM kinase function deregulates the extracellular calcium-sensing receptor (CaR). This malfunction presumably arises from a post-transcriptional event, since CaR mRNA proved to be normal in AT cells. Together, our data suggest that ATM may mediate cell response to mitogenic factors by tightly regulating the set point of the CaR and thereby modulating the crosstalk between this metabotropic receptor and growth factor receptors. Alternatively, the faulty sensing of extracellular calcium in AT cells may be secondary to a state of chronic oxidative stress attributable to ATM deficiency.


Subject(s)
Ataxia Telangiectasia/metabolism , Calcium/metabolism , Fibroblasts/metabolism , Protein Serine-Threonine Kinases/metabolism , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle Proteins , Cells, Cultured , Culture Media, Serum-Free , DNA-Binding Proteins , Enzyme Activation , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptors, Calcium-Sensing , Receptors, Cell Surface/metabolism , Reference Values , Signal Transduction , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...