Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Lab Chip ; 19(2): 343-357, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30566156

ABSTRACT

Cell deformability is a label-free biomarker of cell state in physiological and disease contexts ranging from stem cell differentiation to cancer progression. Harnessing deformability as a phenotype for screening applications requires a method that can simultaneously measure the deformability of hundreds of cell samples and can interface with existing high throughput facilities. Here we present a scalable cell filtration device, which relies on the pressure-driven deformation of cells through a series of pillars that are separated by micron-scale gaps on the timescale of seconds: less deformable cells occlude the gaps more readily than more deformable cells, resulting in decreased filtrate volume which is measured using a plate reader. The key innovation in this method is that we design customized arrays of individual filtration devices in a standard 96-well format using soft lithography, which enables multiwell input samples and filtrate outputs to be processed with higher throughput using automated pipette arrays and plate readers. To validate high throughput filtration to detect changes in cell deformability, we show the differential filtration of human ovarian cancer cells that have acquired cisplatin-resistance, which is corroborated with cell stiffness measurements using quantitative deformability cytometry. We also demonstrate differences in the filtration of human cancer cell lines, including ovarian cancer cells that overexpress transcription factors (Snail, Slug), which are implicated in epithelial-to-mesenchymal transition; breast cancer cells (malignant versus benign); and prostate cancer cells (highly versus weekly metastatic). We additionally show how the filtration of ovarian cancer cells is affected by treatment with drugs known to perturb the cytoskeleton and the nucleus. Our results across multiple cancer cell types with both genetic and pharmacologic manipulations demonstrate the potential of this scalable filtration device to screen cells based on their deformability.


Subject(s)
Cell Separation/instrumentation , Cell Shape/physiology , High-Throughput Screening Assays/instrumentation , Single-Cell Analysis/instrumentation , Cell Line, Tumor , Cell Separation/methods , Humans
2.
Ann Surg Oncol ; 21 Suppl 4: S758-66, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25124473

ABSTRACT

BACKGROUND: Human basal-like breast cancer (BLBC) has a poor prognosis and is often identified by expression of the epidermal growth factor receptor (EGFR). BLBC remains a major clinical challenge because its pathogenesis is not well understood, thus hindering efforts to develop targeted therapies. Recent data implicate the forkhead box C1 (FOXC1) transcription factor as an important prognostic biomarker and functional regulator of BLBC, but its regulatory mechanism and impact on BLBC tumorigenesis remain unclear. METHODS: The association between FOXC1 and EGFR expression in human breast cancer was examined by immunohistochemistry in formalin-fixed tissues and analysis of the TCGA database. The regulation of FOXC1 by EGFR activation was investigated in MDA-MB-468 cells using immunoblotting, qRT-PCR, and luciferase activity assays. This EGFR effect on FOXC1 expression was confirmed using the MDA-MB-468 xenograft model. RESULTS: Both FOXC1 mRNA and protein levels significantly correlated with EGFR expression in human breast tumors. EGFR activation induced FOXC1 transcription through the ERK and Akt pathways in BLBC. EGFR inhibition in vivo reduced FOXC1 expression in xenograft tumors. We also found that FOXC1 knockdown impaired the effects of EGF on BLBC cell proliferation, migration, and invasion. CONCLUSIONS: Our findings uncover a novel EGFR-FOXC1 signaling axis critical for BLBC cell functions, supporting the notion that intervention in the FOXC1 pathway may provide potential modalities for BLBC treatment.


Subject(s)
ErbB Receptors/genetics , Forkhead Transcription Factors/genetics , RNA, Messenger/analysis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement , Cell Proliferation , ErbB Receptors/analysis , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/metabolism , Gefitinib , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/therapeutic use , RNA, Small Interfering/genetics , Transfection , Triple Negative Breast Neoplasms/chemistry , Triple Negative Breast Neoplasms/drug therapy , Up-Regulation
3.
PLoS One ; 7(11): e49466, 2012.
Article in English | MEDLINE | ID: mdl-23166678

ABSTRACT

The phosphoinositide 3-kinase (PI3K) pathway is targeted for frequent alteration in glioblastoma (GBM) and is one of the core GBM pathways defined by The Cancer Genome Atlas. Somatic mutations of PIK3R1 are observed in multiple tumor types, but the tumorigenic activity of these mutations has not been demonstrated in GBM. We show here that somatic mutations in the iSH2 domain of PIK3R1 act as oncogenic driver events. Specifically, introduction of a subset of the mutations identified in human GBM, in the nSH2 and iSH2 domains, increases signaling through the PI3K pathway and promotes tumorigenesis of primary normal human astrocytes in an orthotopic xenograft model. Furthermore, we show that cells that are dependent on mutant P85α-mediated PI3K signaling exhibit increased sensitivity to a small molecule inhibitor of AKT. Together, these results suggest that GBM patients whose tumors carry mutant PIK3R1 alleles may benefit from treatment with inhibitors of AKT.


Subject(s)
Astrocytes/metabolism , Cell Transformation, Neoplastic/genetics , Class Ia Phosphatidylinositol 3-Kinase/genetics , Glioblastoma/genetics , Signal Transduction/genetics , Analysis of Variance , Cell Survival/drug effects , Cell Transformation, Neoplastic/metabolism , Dimethyl Sulfoxide/toxicity , Dose-Response Relationship, Drug , Heterocyclic Compounds, 3-Ring/toxicity , Humans , Immunoblotting , Mutagenesis , Mutation/genetics , Plasmids/genetics
4.
Neuro Oncol ; 14(11): 1325-31, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23074196

ABSTRACT

Large-scale cancer genomics efforts are identifying hundreds of somatic genomic alterations in glioblastoma (GBM). Distinguishing between active driver and neutral passenger alterations requires functional assessment of each gene; therefore, integrating biological weight of evidence with statistical significance for each genomic alteration will enable better prioritization for downstream studies. Here, we demonstrate the feasibility and potential of in vitro functional genomic screens to rapidly and systematically prioritize high-probability candidate genes for in vivo validation. Integration of low-complexity gain- and loss-of-function screens designed on the basis of genomic data identified 6 candidate GBM oncogenes, and RINT1 was validated as a novel GBM oncogene based on its ability to confer tumorigenicity to primary nontransformed murine astrocytes in vivo. Cancer genomics-guided low-complexity genomic screens can quickly provide a functional filter to prioritize high-value targets for further downstream mechanistic and translational studies.


Subject(s)
Brain Neoplasms/genetics , Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic/genetics , Glioblastoma/genetics , Oncogenes/genetics , Animals , Comparative Genomic Hybridization , Genomics , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Nude
5.
Cancer Cell ; 17(5): 497-509, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20478531

ABSTRACT

A hallmark feature of glioblastoma is its strong self-renewal potential and immature differentiation state, which contributes to its plasticity and therapeutic resistance. Here, integrated genomic and biological analyses identified PLAGL2 as a potent protooncogene targeted for amplification/gain in malignant gliomas. Enhanced PLAGL2 expression strongly suppresses neural stem cell (NSC) and glioma-initiating cell differentiation while promoting their self-renewal capacity upon differentiation induction. Transcriptome analysis revealed that these differentiation-suppressive activities are attributable in part to PLAGL2 modulation of Wnt/beta-catenin signaling. Inhibition of Wnt signaling partially restores PLAGL2-expressing NSC differentiation capacity. The identification of PLAGL2 as a glioma oncogene highlights the importance of a growing class of cancer genes functioning to impart stem cell-like characteristics in malignant cells.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/physiology , Glioblastoma/pathology , RNA-Binding Proteins/physiology , Signal Transduction/physiology , Stem Cells/cytology , Transcription Factors/physiology , Wnt Proteins/metabolism , Animals , Cell Transformation, Neoplastic , Humans , Mice
6.
Proc Natl Acad Sci U S A ; 107(15): 6912-7, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20351267

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and lethal primary brain cancer that is driven by aberrant signaling of growth factor receptors, particularly the epidermal growth factor receptor (EGFR). EGFR signaling is tightly regulated by receptor endocytosis and lysosome-mediated degradation, although the molecular mechanisms governing such regulation, particularly in the context of cancer, remain poorly delineated. Here, high-resolution genomic profiles of GBM identified a highly recurrent focal 1p36 deletion encompassing the putative tumor suppressor gene, Mig-6. We show that Mig-6 quells the malignant potential of GBM cells and dampens EGFR signaling by driving EGFR into late endosomes and lysosome-mediated degradation upon ligand stimulation. Mechanistically, this effect is mediated by the binding of Mig-6 to a SNARE protein STX8, a protein known to be required for late endosome trafficking. Thus, Mig-6 functions to ensure recruitment of internalized receptor to late endosomes and subsequently the lysosomal degradation compartment through its ability to specifically link EGFR and STX8 during ligand-stimulated EGFR trafficking. In GBM, the highly frequent loss of Mig-6 would therefore serve to sustain aberrant EGFR-mediated oncogenic signaling. Together, these data uncover a unique tumor suppression mechanism involving the regulation of receptor trafficking.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Brain Neoplasms/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Tumor Suppressor Proteins/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Animals , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Humans , Intracellular Signaling Peptides and Proteins , Lysosomes/metabolism , Mice , Neoplasm Invasiveness , Tumor Suppressor Proteins/genetics , Two-Hybrid System Techniques
7.
Nature ; 455(7216): 1129-33, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18948956

ABSTRACT

Glioblastoma (GBM) is a highly lethal brain tumour presenting as one of two subtypes with distinct clinical histories and molecular profiles. The primary GBM subtype presents acutely as a high-grade disease that typically harbours mutations in EGFR, PTEN and INK4A/ARF (also known as CDKN2A), and the secondary GBM subtype evolves from the slow progression of a low-grade disease that classically possesses PDGF and TP53 events. Here we show that concomitant central nervous system (CNS)-specific deletion of p53 and Pten in the mouse CNS generates a penetrant acute-onset high-grade malignant glioma phenotype with notable clinical, pathological and molecular resemblance to primary GBM in humans. This genetic observation prompted TP53 and PTEN mutational analysis in human primary GBM, demonstrating unexpectedly frequent inactivating mutations of TP53 as well as the expected PTEN mutations. Integrated transcriptomic profiling, in silico promoter analysis and functional studies of murine neural stem cells (NSCs) established that dual, but not singular, inactivation of p53 and Pten promotes an undifferentiated state with high renewal potential and drives increased Myc protein levels and its associated signature. Functional studies validated increased Myc activity as a potent contributor to the impaired differentiation and enhanced renewal of NSCs doubly null for p53 and Pten (p53(-/-) Pten(-/-)) as well as tumour neurospheres (TNSs) derived from this model. Myc also serves to maintain robust tumorigenic potential of p53(-/-) Pten(-/-) TNSs. These murine modelling studies, together with confirmatory transcriptomic/promoter studies in human primary GBM, validate a pathogenetic role of a common tumour suppressor mutation profile in human primary GBM and establish Myc as an important target for cooperative actions of p53 and Pten in the regulation of normal and malignant stem/progenitor cell differentiation, self-renewal and tumorigenic potential.


Subject(s)
Brain Neoplasms/pathology , Cell Differentiation , Glioma/pathology , Neoplastic Stem Cells/pathology , Neurons/pathology , PTEN Phosphohydrolase/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Brain Neoplasms/genetics , Cell Proliferation , Gene Expression Regulation , Glioblastoma/genetics , Glioblastoma/pathology , Glioma/genetics , Humans , Immunohistochemistry , Mice , Neoplastic Stem Cells/metabolism , Neurons/metabolism , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Protein p53/genetics
8.
Cancer Cell ; 13(4): 355-64, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18394558

ABSTRACT

We have developed a nonheuristic genome topography scan (GTS) algorithm to characterize the patterns of genomic alterations in human glioblastoma (GBM), identifying frequent p18(INK4C) and p16(INK4A) codeletion. Functional reconstitution of p18(INK4C) in GBM cells null for both p16(INK4A) and p18(INK4C) resulted in impaired cell-cycle progression and tumorigenic potential. Conversely, RNAi-mediated depletion of p18(INK4C) in p16(INK4A)-deficient primary astrocytes or established GBM cells enhanced tumorigenicity in vitro and in vivo. Furthermore, acute suppression of p16(INK4A) in primary astrocytes induced a concomitant increase in p18(INK4C). Together, these findings uncover a feedback regulatory circuit in the astrocytic lineage and demonstrate a bona fide tumor suppressor role for p18(INK4C) in human GBM wherein it functions cooperatively with other INK4 family members to constrain inappropriate proliferation.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Feedback, Physiological , Glioblastoma/metabolism , Glioblastoma/pathology , Animals , Astrocytes/pathology , Cell Line, Tumor , Cells, Cultured , Gene Deletion , Gene Dosage , Gene Expression Regulation, Neoplastic , Genome, Human , Glioblastoma/genetics , Humans , Mice , Up-Regulation/genetics
9.
Science ; 318(5848): 287-90, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17872411

ABSTRACT

Targeted therapies that inhibit receptor tyrosine kinases (RTKs) and the downstream phosphatidylinositol 3-kinase (PI3K) signaling pathway have shown promising anticancer activity, but their efficacy in the brain tumor glioblastoma multiforme (GBM) and other solid tumors has been modest. We hypothesized that multiple RTKs are coactivated in these tumors and that redundant inputs drive and maintain downstream signaling, thereby limiting the efficacy of therapies targeting single RTKs. Tumor cell lines, xenotransplants, and primary tumors indeed show multiple concomitantly activated RTKs. Combinations of RTK inhibitors and/or RNA interference, but not single agents, decreased signaling, cell survival, and anchorage-independent growth even in glioma cells deficient in PTEN, a frequently inactivated inhibitor of PI3K. Thus, effective GBM therapy may require combined regimens targeting multiple RTKs.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/enzymology , Glioblastoma/enzymology , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival , Enzyme Activation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Glioblastoma/drug therapy , Humans , Indoles/pharmacology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Piperazines/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-met , Quinazolines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/metabolism , Signal Transduction , Sulfonamides/pharmacology
10.
Clin Cancer Res ; 13(16): 4695-703, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17652624

ABSTRACT

PURPOSE: Amplified MYCN oncogene defines a subgroup of neuroblastomas with poor outcome. However, a substantial number of MYCN single-copy neuroblastomas exhibits an aggressive phenotype similar to that of MYCN-amplified neuroblastomas even in the absence of high MYCN mRNA and/or protein levels. EXPERIMENTAL DESIGN: To identify shared molecular mechanisms that mediate the aggressive phenotype in MYCN-amplified and single-copy high-risk neuroblastomas, we defined genetic programs evoked by ectopically expressed MYCN in vitro and analyzed them in high-risk versus low-risk neuroblastoma tumors (n = 49) using cDNA microarrays. Candidate gene expression was validated in a separate cohort of 117 patients using quantitative PCR, and protein expression was analyzed in neuroblastoma tumors by immunoblotting and immunohistochemistry. RESULTS: We identified a genetic signature characterized by a subset of MYCN/MYC and E2F targets, including Skp2, encoding the F-box protein of the SCF(Skp2) E3-ligase, to be highly expressed in high-risk neuroblastomas independent of amplified MYCN. We validated the findings for Skp2 and analyzed its expression in relation to MYCN and E2F-1 expression in a separate cohort (n = 117) using quantitative PCR. High Skp2 expression proved to be a highly significant marker of dire prognosis independent of both MYCN status and disease stage, on the basis of multivariate analysis of event-free survival (hazard ratio, 3.54; 95% confidence interval, 1.56-8.00; P = 0.002). Skp2 protein expression was inversely correlated with expression of p27, the primary target of the SCF(Skp2) E3-ligase, in neuroblastoma tumors. CONCLUSION: Skp2 may have a key role in the progression of neuroblastomas and should make an attractive target for therapeutic approaches.


Subject(s)
Genes, myc , Neuroblastoma/genetics , S-Phase Kinase-Associated Proteins/physiology , E2F1 Transcription Factor/genetics , Gene Expression Profiling , Humans , Immunohistochemistry , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , Risk , S-Phase Kinase-Associated Proteins/analysis , S-Phase Kinase-Associated Proteins/genetics
11.
Nature ; 447(7147): 966-71, 2007 Jun 21.
Article in English | MEDLINE | ID: mdl-17515920

ABSTRACT

Highly rearranged and mutated cancer genomes present major challenges in the identification of pathogenetic events driving the neoplastic transformation process. Here we engineered lymphoma-prone mice with chromosomal instability to assess the usefulness of mouse models in cancer gene discovery and the extent of cross-species overlap in cancer-associated copy number aberrations. Along with targeted re-sequencing, our comparative oncogenomic studies identified FBXW7 and PTEN to be commonly deleted both in murine lymphomas and in human T-cell acute lymphoblastic leukaemia/lymphoma (T-ALL). The murine cancers acquire widespread recurrent amplifications and deletions targeting loci syntenic to those not only in human T-ALL but also in diverse human haematopoietic, mesenchymal and epithelial tumours. These results indicate that murine and human tumours experience common biological processes driven by orthologous genetic events in their malignant evolution. The highly concordant nature of genomic events encourages the use of genomically unstable murine cancer models in the discovery of biological driver events in the human oncogenome.


Subject(s)
Chromosomal Instability/genetics , Chromosome Aberrations , Conserved Sequence/genetics , Leukemia-Lymphoma, Adult T-Cell/genetics , Lymphoma, T-Cell/genetics , Animals , Genome/genetics , Humans , Mice , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Synteny/genetics
12.
Cancer Lett ; 243(1): 90-100, 2006 Nov 08.
Article in English | MEDLINE | ID: mdl-16448746

ABSTRACT

The BRCA2 protein is involved in the maintenance of genomic stability through its key role in homologous recombination repair of DNA double strand breaks. Biallelic inactivation of BRCA2 leads to a defect in DNA repair and is associated with a chromosomal instability phenotype. Recent studies on familial breast cancer clusters revealed chromosomal rearrangements and higher rates of sister chromatid exchanges also in heterozygous BRCA2 mutation carriers. In the present study, lymphoblastoid cell lines of heterozygous BRCA2 mutation carriers and of wildtype relatives were compared with regard to BRCA2 mRNA and protein expression and capacity to repair DNA damage induced by gamma-irradiation and mitomycin C. BRCA2+/- cells showed lower amounts of the full-length BRCA2 protein compared to BRCA2+/+ cells. The kinetics of gamma-H2AX protein level revealed distinct defects in DNA double strand break repair in the BRCA2+/- cells. These results are indicative of a haploinsufficiency phenotype in BRCA2+/- cells, suggesting that reduced amounts of functional BRCA2 protein in BRCA2+/- carriers are insufficient for an efficient repair of DNA double strand breaks, a condition that could contribute to the impairment of genomic stability.


Subject(s)
BRCA2 Protein/metabolism , DNA Damage/genetics , DNA Repair/genetics , Mutation/genetics , Analysis of Variance , BRCA2 Protein/genetics , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms, Male/genetics , Breast Neoplasms, Male/metabolism , Breast Neoplasms, Male/pathology , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line , Comet Assay , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Female , Flow Cytometry , Fluorescent Antibody Technique , Gamma Rays , Haplotypes/genetics , Heterozygote , Humans , Male , Mitomycin/pharmacology , Mutation/drug effects , Mutation/radiation effects , Pedigree , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
Clin Cancer Res ; 12(1): 131-8, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16397034

ABSTRACT

PURPOSE: A distal portion of 1p is frequently deleted in human neuroblastomas, and it is generally assumed that this region harbors at least one gene relevant for neuroblastoma development. A 1p36.3 commonly deleted region, bordered by D1S2731 and D1S214 has been defined. The present study surveys whether expression of genes mapping to this region is associated with tumor behavior. EXPERIMENTAL DESIGN: Candidate genes localized within the deleted region were identified by sequence data analysis. Their expression was assessed in a cohort of 49 primary neuroblastomas using cDNA microarray analysis. Gene expression patterns associated with known prognostic markers and patient outcome were further evaluated by quantitative real-time reverse transcription-PCR in a cohort of 102 neuroblastomas. RESULTS: The commonly deleted region spans 261 kb and encompasses two genes, FLJ10737 and CAMTA1. We found no evidence for an association of FLJ10737 expression with established prognostic variables or outcome. In contrast, low CAMTA1 expression characterized tumors with 1p deletion, MYCN amplification, and advanced tumor stages 3 and 4. Moreover, low CAMTA1 expression was significantly associated with poor outcome (P < 0.001). In multivariate analysis of event-free survival, the prognostic information of low CAMTA1 expression was independent of 1p status, MYCN status, tumor stage, and age of the patient at diagnosis (hazard ratio, 3.52; 95% confidence interval, 1.21-10.28; P = 0.02). CONCLUSIONS: Our data suggest that assessment of CAMTA1 expression may improve the prognostic models for neuroblastoma and that it will be important to define the biological function of CAMTA1 in this disease.


Subject(s)
Biomarkers, Tumor/analysis , Calcium-Binding Proteins/biosynthesis , Chromosomes, Human, Pair 1/genetics , Neuroblastoma/metabolism , Neuroblastoma/mortality , Trans-Activators/biosynthesis , Age Factors , Calcium-Binding Proteins/genetics , Humans , Infant , Neuroblastoma/pathology , Oligonucleotide Array Sequence Analysis , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Trans-Activators/genetics
14.
Cancer Res ; 63(22): 7742-52, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14633699

ABSTRACT

In neuroblastoma cells, apoptotic programs can be activated by cytokines and cytostatic drugs. Apoptotic dysfunction confers resistance against therapeutic drugs and is a major complication for achieving optimal therapy response. Deregulated expression of the MYCN gene is a critical determinant in neuroblastoma progression, and one of the pleiotropic functions of the MYCN protein is cellular sensitization to cytokine-induced and drug-induced apoptosis. By using the functional approach of technical knockout (TKO), we have identified five genes that regulate sensitization for IFN-gamma-induced cell death. Most efficient among them is the newly identified SOXN (neuroblastoma-derived sulfhydryl oxidase), which comprises 12 exons and maps to 9q34.3. SOXN encodes a putative protein of 698 amino acids that contains a signal sequence, a protein-disulfide-isomerase-type thioredoxin and a yeast ERV1 domain and is highly homologous to members of the sulfhydryl oxidase/Quiescin6 family. The SOXN protein is predominantly located in the plasma and in the nuclear membrane. Antisense SOXN confers resistance to IFN-gamma-induced apoptosis. In contrast, ectopic overexpression of sense-SOXN sensitizes the cells to induced cell death. These results identify SOXN as a major player in regulating the sensitization of neuroblastoma cells for IFN-gamma-induced apoptosis.


Subject(s)
Apoptosis/physiology , Interferon-gamma/pharmacology , Neuroblastoma/enzymology , Oxidoreductases/physiology , Amino Acid Sequence , Apoptosis/drug effects , Apoptosis/genetics , Base Sequence , Cell Membrane/enzymology , Chromosomes, Human, Pair 9/genetics , DNA, Antisense/genetics , DNA, Complementary/genetics , Exons , Humans , Molecular Sequence Data , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/pathology , Oxidoreductases/genetics , Oxidoreductases/metabolism , Oxidoreductases Acting on Sulfur Group Donors , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sequence Homology, Amino Acid , Transfection
15.
Oncogene ; 22(3): 401-11, 2003 Jan 23.
Article in English | MEDLINE | ID: mdl-12545161

ABSTRACT

Neuroblastoma is a highly heterogeneous tumor of young children. Although many advances have been made towards understanding the molecular mechanisms dictating the phenotypic heterogeneity, the prognosis of children with neuroblastoma, particularly of progressively growing variants, has remained dire. About 10% of neuroblastomas regress spontaneously, probably by apoptosis, while another 20% have amplified the MYCN gene resulting in a poor prognosis. In pursuit of identifying cell death-associated genes in neuroblastoma, we encountered the SCA2 gene, coding for ataxin-2, as an important player. Here, we report that enforced expression of wild-type ataxin-2, but not of mutant ataxin-2, sensitizes neuroblastoma cells for apoptosis. In line with this, higher levels of ataxin-2 were detected in apoptotic cells compared to nonapoptotic cells. Neuroblastoma tumors with amplified MYCN contain significantly less ataxin-2 protein than tumors without amplified MYCN. Collectively, our data suggest that ataxin-2 has an important role in regulating the susceptibility of neuroblastoma cells to apoptotic stimuli in vitro and in vivo.


Subject(s)
Apoptosis/physiology , Neuroblastoma/pathology , Proteins/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Ataxins , Caspase Inhibitors , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mutation , N-Myc Proto-Oncogene Protein , Nerve Tissue Proteins , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Peptides/genetics , Proteins/genetics , Tumor Cells, Cultured , Up-Regulation
16.
Cancer Cell ; 2(5): 377-86, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12450793

ABSTRACT

Tumor stage, age of patient, and amplification of MYCN predict disease outcome in neuroblastoma. To gain insight into the underlying molecular pathways, we have obtained expression profiles from 94 primary neuroblastoma specimens. Advanced tumor stages show a characteristic expression profile that includes downregulation of multiple genes involved in signal transduction through Fyn and the actin cytoskeleton. High expression of Fyn and high Fyn kinase activity are restricted to low-stage tumors. In culture, expression of active Fyn kinase induces differentiation and growth arrest of neuroblastoma cells. Expression of Fyn predicts long-term survival independently of MYCN amplification. Amplification of MYCN correlates with deregulation of a distinct set of genes, many of which are target genes of Myc. Our data demonstrate a causal role for Fyn kinase in the genesis of neuroblastoma.


Subject(s)
Gene Expression Regulation, Neoplastic , Neuroblastoma/genetics , Proto-Oncogene Proteins/metabolism , Cell Differentiation/genetics , Cell Division/genetics , Disease-Free Survival , Gene Expression Profiling , Genes, myc , Humans , Infant , N-Myc Proto-Oncogene Protein , Neuroblastoma/metabolism , Neuroblastoma/mortality , Neuroblastoma/pathology , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Oncogene Proteins/metabolism , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-fyn , Survival Analysis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...