Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Immunol ; 196(4): 1910-21, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26792806

ABSTRACT

Osteoarthritis is a degenerative joint disease that ranks among the leading causes of adult disability. Mechanisms underlying osteoarthritis pathogenesis are not yet fully elucidated, putting limits to current disease management and treatment. Based on the phenomenological evidence for dysregulation within the glycome of chondrocytes and the network of a family of adhesion/growth-regulatory lectins, that is, galectins, we tested the hypothesis that Galectin-1 is relevant for causing degeneration. Immunohistochemical analysis substantiated that Galectin-1 upregulation is associated with osteoarthritic cartilage and subchondral bone histopathology and severity of degeneration (p < 0.0001, n = 29 patients). In vitro, the lectin was secreted and it bound to osteoarthritic chondrocytes inhibitable by cognate sugar. Glycan-dependent Galectin-1 binding induced a set of disease markers, including matrix metalloproteinases and activated NF-κB, hereby switching on an inflammatory gene signature (p < 10(-16)). Inhibition of distinct components of the NF-κB pathway using dedicated inhibitors led to dose-dependent impairment of Galectin-1-mediated transcriptional activation. Enhanced secretion of effectors of degeneration such as three matrix metalloproteinases underscores the data's pathophysiological relevance. This study thus identifies Galectin-1 as a master regulator of clinically relevant inflammatory-response genes, working via NF-κB. Because inflammation is critical to cartilage degeneration in osteoarthritis, this report reveals an intimate relation of glycobiology to osteoarthritic cartilage degeneration.


Subject(s)
Galectin 1/metabolism , Gene Expression Regulation/physiology , Gene Regulatory Networks/physiology , NF-kappa B/metabolism , Osteoarthritis/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Glycomics , Humans , Immunohistochemistry , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Osteoarthritis/genetics , Osteoarthritis/pathology , Reverse Transcriptase Polymerase Chain Reaction
2.
Int J Cancer ; 138(5): 1220-31, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26414866

ABSTRACT

Transmembrane tyrosine-kinase Ephrin receptors promote tumor progression and/or metastasis of several malignancies including leukemia, follicular lymphoma, glioma, malignant pleural mesothelioma, papillary thyroid carcinoma, sarcomas and ovarian, breast, bladder and non-small cell lung cancers. They also drive intestinal stem cell proliferation and positioning, control intestinal tissue boundaries and are involved in liver, pancreatic and colorectal cancers, indicating involvement in additional digestive system malignancies. We investigated the role of Ephrin-B4 receptor (EPHB4), and its ligand EFNB2, in gastric and gastroesophageal junction cancers in patient cohorts through computational, mathematical, molecular and immunohistochemical analyses. We show that EPHB4 is upregulated in preneoplastic gastroesophageal lesions and its expression further increased in gastroesophageal cancers in several independent cohorts. The closely related EPHB6 receptor, which also binds EFNB2, was downregulated in all tested cohorts, consistent with its tumor-suppressive properties in other cancers. EFNB2 expression is induced in esophageal cells by acidity, suggesting that gastroesophageal reflux disease (GERD) may constitute an early triggering event in activating EFNB2-EPHB4 signaling. Association of EPHB4 to both Barrett's esophagus and to advanced tumor stages, and its overexpression at the tumor invasion front and vascular endothelial cells intimate the notion that EPHB4 may be associated with multiple steps of gastroesophageal tumorigenesis. Analysis of oncogenomic signatures uncovered the first EPHB4-associated gene network (false discovery rate: 7 × 10(-90) ) composed of a five-transcription factor interconnected gene network that drives proliferation, angiogenesis and invasiveness. The EPHB4 oncogenomic network provides a molecular basis for its role in tumor progression and points to EPHB4 as a potential tumor aggressiveness biomarker and drug target in gastroesophageal cancers.


Subject(s)
Esophageal Neoplasms/etiology , Esophagogastric Junction , Gene Regulatory Networks , Receptor, EphB4/physiology , Stomach Neoplasms/etiology , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Ephrin-B2/physiology , Esophageal Neoplasms/genetics , Female , Humans , Male , Middle Aged , Receptor Protein-Tyrosine Kinases/physiology , Receptor, EphB4/analysis , Receptor, EphB4/genetics , Receptors, Eph Family , Stomach Neoplasms/genetics
3.
Cancer Inform ; 14: 131-9, 2015.
Article in English | MEDLINE | ID: mdl-26568679

ABSTRACT

Whole-genome analyses have uncovered that most cancer-relevant genes cluster into 12 signaling pathways. Knowledge of the signaling pathways and associated gene signatures not only allows us to understand the mechanisms of oncogenesis inherent to specific cancers but also provides us with drug targets, molecular diagnostic and prognosis factors, as well as biomarkers for patient risk stratification and treatment. Publicly available genomic data sets constitute a wealth of gene mining opportunities for hypothesis generation and testing. However, the increasingly recognized genetic and epigenetic inter- and intratumor heterogeneity, combined with the preponderance of small-size cohorts, hamper reliable analysis and discovery. Here, we review two methods that are used to infer meaningful biological events from small-size data sets and discuss some of their applications and limitations.

4.
PLoS One ; 10(9): e0139101, 2015.
Article in English | MEDLINE | ID: mdl-26406496

ABSTRACT

Endothelial cells are often present at inflammation sites. This is the case of endothelial cells of the blood-brain barrier (BBB) of patients afflicted with neurodegenerative disorders such as Alzheimer's, Parkinson's, or multiple sclerosis, as well as in cases of bacterial meningitis, trauma, or tumor-associated ischemia. Inflammation is a known modulator of gene expression through the activation of transcription factors, mostly NF-κB. RLIP76 (a.k.a. RALBP1), an ATP-dependent transporter of electrophile-glutathione conjugates, modulates BBB permeability through the regulation of tight junction function, cell adhesion, and exocytosis. Genes and pathways regulated by RLIP76 are transcriptional targets of tumor necrosis factor alpha (TNF-α) pro-inflammatory molecule, suggesting that RLIP76 may also be an inflammation target. To assess the effects of TNF-α on RLIP76, we faced the problem of choosing reference genes impervious to TNF-α. Since such genes were not known in human BBB endothelial cells, we subjected these to TNF-α, and measured by quantitative RT-PCR the expression of housekeeping genes commonly used as reference genes. We find most to be modulated, and analysis of several inflammation datasets as well as a metaanalysis of more than 5000 human tissue samples encompassing more than 300 cell types and diseases show that no single housekeeping gene may be used as a reference gene. Using three different algorithms, however, we uncovered a reference geneset impervious to TNF-α, and show for the first time that RLIP76 expression is induced by TNF-α and follows the induction kinetics of inflammation markers, suggesting that inflammation can influence RLIP76 expression at the BBB. We also show that MRP1 (a.k.a. ABCC1), another electrophile-glutathione transporter, is not modulated in the same cells and conditions, indicating that RLIP76 regulation by TNF-α is not a general property of glutathione transporters. The reference geneset uncovered herein should aid in future gene expression studies in inflammatory conditions of the BBB.


Subject(s)
Blood-Brain Barrier/pathology , Endothelial Cells/metabolism , Genes, Essential , Glutathione/metabolism , Inflammation/genetics , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Binding Sites , Biomarkers/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Expression Regulation/drug effects , Genetic Association Studies , Humans , Inflammation/pathology , Mice , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , Rats , Software , Tumor Necrosis Factor-alpha/pharmacology
5.
Breast Cancer Res ; 14(5): 112, 2012 Sep 17.
Article in English | MEDLINE | ID: mdl-22992372

ABSTRACT

LSD1, an epigenetic modifier, and PELP1, an estrogen receptor co-activator, integrate estrogen receptor ERα and HER2 receptor tyrosine kinase signaling to promote aromatase expression and hormone resistance in a preclinical model of post-menopausal breast cancer. In the previous issue of Breast Cancer Research, Cortez et al. show, for the first time, that knockdown or drug-mediated inhibition of PELP1 or LSD1 suppresses LSD1-mediated transcriptionally activating histone marks at ERα target genes, inhibits aromatase gene expression, and sensitizes hormone refractory breast cancer cells to tamoxifen or letrozole treatments. The relevance of PELP1-LSD1 signaling to other nuclear hormone receptor-dependent cancers and structural considerations for the selective drug targeting of LSD1 are further discussed in this editorial.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Co-Repressor Proteins/genetics , Histone Demethylases/metabolism , Transcription Factors/genetics , Animals , Female , Humans
6.
Hum Pathol ; 43(8): 1300-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22245111

ABSTRACT

Lysine-specific demethylase 1 (GeneID 23028), a flavin-dependent monoamine oxidoreductase and a histone demethylase, serves as an epigenetic coregulator of transcription. Lysine-specific demethylase 1 is up-regulated in neuroblastoma and in bladder, breast, colorectal, gastric, lung, and neuroendocrine cancers, and its overexpression drives the cell cycle of otherwise nontransformed human cells, suggesting oncogenic properties. Lysine-specific demethylase 1 was recently reported to be also overexpressed in several different mesenchymal tumors. We investigated lysine-specific demethylase 1 expression in over 500 sarcomas by gene expression profiling and tissue microarray-coupled immunohistochemical analyses and confirmed lysine-specific demethylase 1 overexpression in rhabdomyosarcoma and synovial sarcoma. We also show for the first time that lysine-specific demethylase 1 is also overexpressed in chondrosarcoma, Ewing's sarcoma, and osteosarcoma wherein it localizes in cell nuclei. We further show that a US Food and Drug Administration-approved drug that inhibits lysine-specific demethylase 1 also inhibits chondrosarcoma, Ewing's sarcoma, osteosarcoma, and rhabdomyosarcoma cell growth in vitro. These data suggest that lysine-specific demethylase 1 plays a role in sarcoma pathology and that lysine-specific demethylase 1 inhibition strategies might represent a novel means to inhibiting growth of lysine-specific demethylase 1-overexpressing sarcomas.


Subject(s)
Bone Neoplasms/genetics , Chondrosarcoma/genetics , Histone Demethylases/genetics , Osteosarcoma/genetics , Rhabdomyosarcoma/genetics , Sarcoma, Ewing/genetics , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Cell Proliferation/drug effects , Chondrosarcoma/drug therapy , Chondrosarcoma/pathology , Gene Expression Profiling , Histone Demethylases/antagonists & inhibitors , Humans , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/pathology , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/pathology , Tranylcypromine/pharmacology , Tranylcypromine/therapeutic use
7.
Gene ; 491(2): 103-9, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22019431

ABSTRACT

Several gene databases, including heavily used ones such as the National Center for Biotechnology Information (NCBI) database, erroneously assign, on occasion, literature references to genes or proteins. These mistakes are mostly due to an overlap in gene aliases, whereby two distinct genes share a pseudonym. This is particularly confusing when the gene products have also biological properties in common, are part of signaling pathways that cross-talk to one another, or are regulated by the same effectors. We present examples spanning several research fields including apoptosis, ubiquitin-dependent degradation, signaling by Notch, Wnt, and small G proteins, transporters of glutathione conjugates of electrophiles, and mitochondrial and ribosomal RNA genes. To solve the problem, we argue in favor of including Entrez gene numbers in papers submitted for publication as unique gene identifiers to allow precise identification of genes and species studied.


Subject(s)
Databases, Genetic , Genes , Terminology as Topic , Animals , Genomics , National Library of Medicine (U.S.) , United States
8.
Epigenomics ; 3(6): 715-32, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22126291

ABSTRACT

Sarcomas comprise a large number of rare, histogenetically heterogeneous, mesenchymal tumors. Cancers such as Ewing's sarcoma, liposarcoma, rhabdomyosarcoma and synovial sarcoma can be generated by the transduction of mesenchymal stem cell progenitors with sarcoma-pathognomonic oncogenic fusions, a neoplastic transformation process accompanied by profound locus-specific and pangenomic epigenetic alterations. The epigenetic activities of histone-modifying and chromatin-remodeling enzymes such as SUV39H1/KMT1A, EZH2/KMT6A and BMI1 are central to epigenetic-regulated transformation, a property we coin oncoepigenic. Sarcoma-specific oncoepigenic aberrations modulate critical signaling pathways that control cell growth and differentiation including several miRNAs, Wnt, PI3K/AKT, Sav-RASSF1-Hpo and regulators of the G1 and G2/M checkpoints of the cell cycle. Herein an overview of the current knowledge of this rapidly evolving field that will undoubtedly uncover additional oncoepigenic mechanisms and yield druggable targets in the near future is discussed.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epigenesis, Genetic/physiology , Mesenchymal Stem Cells/metabolism , Models, Biological , Sarcoma/physiopathology , Animals , Genes, Tumor Suppressor/physiology , Histone Demethylases/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Humans , Mice , MicroRNAs/metabolism , Sarcoma/metabolism
9.
J Pathol ; 225(3): 353-63, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21984123

ABSTRACT

Notch can act as an oncogene or as a tumour suppressor and thus can either promote or inhibit tumour cell growth. To establish Notch status in Ewing's sarcoma family of tumours (ESFT), we investigated the Notch pathway by gene expression profiling meta-analysis or immunohistochemistry in samples obtained from 96 and 24 ESFT patients, respectively. We found that although Notch receptors were highly expressed, Notch did not appear to be active, as evidenced by the absence of Notch receptors in cell nuclei. In contrast, we show that Notch receptors known to be active in colon adenocarcinoma, hepatocarcinoma, and pancreatic carcinoma stain cell nuclei in these tumours. High expression of the Notch effector HES1 transcription factor, usually used as a surrogate marker for active Notch, was also restricted to outside of the nucleus in the majority of ESFT, and analysis of HES1 gene targets indicated HES1 to be transcriptionally inactive. Neither forced activation nor pharmacological or genetic blocking of Notch affected HES1 expression in ESFT cells, indicating HES1 expression to be uncoupled from the Notch pathway. Additional functional studies in ESFT cell lines confirmed Notch to be switched off. Finally, unlike experiments in which HES1 expression was modulated, experimental activation of Notch in ESFT cell lines via several means blocked cell proliferation and reduced their clonogenic potential in soft agar. These indicate that HES1 is uncoupled from Notch in ESFT, that EWS-FLI1-mediated inhibition of Notch contributes to ESFT aggressive cell growth, and support a role for Notch in ESFT tumour suppression, at least partly through the Notch effector HEY1.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Neoplasms/metabolism , Homeodomain Proteins/metabolism , Receptors, Notch/metabolism , Sarcoma, Ewing/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Nucleus/metabolism , Cell Proliferation , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/pathology , Receptors, Notch/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Signal Transduction/physiology , Transcription Factor HES-1 , Tumor Cells, Cultured
10.
Clin Cancer Res ; 16(14): 3769-78, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20525755

ABSTRACT

PURPOSE: We report a novel analytic method, named intercohort co-analysis or Ican, which aids in the discovery of genes with predictive value for the progression or outcome of diseases from small-size cohorts. We tested this premise in Ewing's sarcoma (ES), a highly metastatic cancer of bone and soft tissues that lacks validated molecular metastasis and prognostic indicators. EXPERIMENTAL DESIGN: To uncover genes significantly expressed in ES patient subsets, we first determined a nonarbitrary gene expression significance cutoff based on expression levels in validated expressing and nonexpressing tissues. We next searched for genes that were consistently significantly expressed in several ES cohort and cell line datasets. Significantly expressed genes were independently validated by quantitative reverse transcription-PCR in an additional ES cohort. RESULTS: Analysis of ES cohorts revealed marked intercohort gene expression variability. After filtering out the intercohort variability, CXCR4 and CXCR7 were found to be consistently associated with specific ES subsets. Pairwise analyses showed CXCR4 to correlate with ES metastases, and CXCR4 and CXCR7 to patient survival, but not with several other clinicopathological variables. CONCLUSION: Ican is a powerful novel method to identifying genes consistently associated with particular disease states in cancers for which large cohorts are not available, currently the case of most cancers. We report for the first time that high CXCR4 expression preferentially associates with metastatic ES, and that of CXCR7 with poor patient survival.


Subject(s)
Bone Neoplasms/diagnosis , Bone Neoplasms/genetics , Gene Expression Profiling/methods , Receptors, CXCR4/genetics , Receptors, CXCR/genetics , Sarcoma, Ewing/diagnosis , Sarcoma, Ewing/genetics , Bone Neoplasms/pathology , Cohort Studies , Humans , Phenotype , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Ewing/pathology , Tumor Cells, Cultured
11.
Cancer Res ; 70(10): 4015-23, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20442286

ABSTRACT

Hypoxia is an important condition in the tumor cell microenvironment and approximately 1% to 1.5% of the genome is transcriptionally responsive to hypoxia with hypoxia-inducible factor-1 (HIF-1) as a major mediator of transcriptional activation. Tumor hypoxia is associated with a more aggressive phenotype of many cancers in adults, but data on pediatric tumors are scarce. Because, by immunohistochemistry, HIF-1alpha expression was readily detectable in 18 of 28 primary Ewing's sarcoma family tumors (ESFT), a group of highly malignant bone-associated tumors in children and young adults, we studied the effect of hypoxia on ESFT cell lines in vitro. Intriguingly, we found that EWS-FLI1 protein expression, which characterizes ESFT, is upregulated by hypoxia in a HIF-1alpha-dependent manner. Hypoxia modulated the EWS-FLI1 transcriptional signature relative to normoxic conditions. Both synergistic as well as antagonistic transcriptional effects of EWS-FLI1 and of hypoxia were observed. Consistent with alterations in the expression of metastasis-related genes, hypoxia stimulated the invasiveness and soft agar colony formation of ESFT cells in vitro. Our data represent the first transcriptome analysis of hypoxic ESFT cells and identify hypoxia as an important microenvironmental factor modulating EWS-FLI1 expression and target gene activity with far-reaching consequences for the malignant properties of ESFT.


Subject(s)
Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Gene Expression Regulation, Neoplastic/physiology , Hypoxia/metabolism , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , Sarcoma, Ewing/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Profiling , Gene Regulatory Networks , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoenzyme Techniques , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Protein EWS , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Ewing/pathology , Transcriptional Activation
12.
Proc Natl Acad Sci U S A ; 106(13): 5324-9, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19289832

ABSTRACT

Ewing tumors (ET) are highly malignant, localized in bone or soft tissue, and are molecularly defined by ews/ets translocations. DNA microarray analysis revealed a relationship of ET to both endothelium and fetal neural crest. We identified expression of histone methyltransferase enhancer of Zeste, Drosophila, Homolog 2 (EZH2) to be increased in ET. Suppressive activity of EZH2 maintains stemness in normal and malignant cells. Here, we found EWS/FLI1 bound to the EZH2 promoter in vivo, and induced EZH2 expression in ET and mesenchymal stem cells. Down-regulation of EZH2 by RNA interference in ET suppressed oncogenic transformation by inhibiting clonogenicity in vitro. Similarly, tumor development and metastasis was suppressed in immunodeficient Rag2(-/-)gamma(C)(-/-) mice. EZH2-mediated gene silencing was shown to be dependent on histone deacetylase (HDAC) activity. Subsequent microarray analysis of EZH2 knock down, HDAC-inhibitor treatment and confirmation in independent assays revealed an undifferentiated phenotype maintained by EZH2 in ET. EZH2 regulated stemness genes such as nerve growth factor receptor (NGFR), as well as genes involved in neuroectodermal and endothelial differentiation (EMP1, EPHB2, GFAP, and GAP43). These data suggest that EZH2 might have a central role in ET pathology by shaping the oncogenicity and stem cell phenotype of this tumor.


Subject(s)
DNA-Binding Proteins/physiology , Endothelial Cells/pathology , Neural Plate/pathology , Sarcoma, Ewing/etiology , Transcription Factors/physiology , Animals , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Histone Deacetylases , Humans , Mesenchymal Stem Cells , Mice , Neoplasm Metastasis , Oncogene Proteins, Fusion , Polycomb Repressive Complex 2 , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Sarcoma, Ewing/pathology
13.
Cancer Res ; 68(17): 7100-9, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18757425

ABSTRACT

Although p53 is the most frequently mutated gene in cancer, half of human tumors retain wild-type p53, whereby it is unknown whether normal p53 function is compromised by other cancer-associated alterations. One example is Ewing's sarcoma family tumors (ESFT), where 90% express wild-type p53. ESFT are characterized by EWS-FLI1 oncogene fusions. Studying 6 ESFT cell lines, silencing of EWS-FLI1 in a wild-type p53 context resulted in increased p53 and p21(WAF1/CIP1) levels, causing cell cycle arrest. Using a candidate gene approach, HEY1 was linked to p53 induction. HEY1 was rarely expressed in 59 primary tumors, but consistently induced upon EWS-FLI1 knockdown in ESFT cell lines. The NOTCH signaling pathway targets HEY1, and we show NOTCH2 and NOTCH3 to be expressed in ESFT primary tumors and cell lines. Upon EWS-FLI1 silencing, NOTCH3 processing accompanied by nuclear translocation of the activated intracellular domain was observed in all but one p53-mutant cell line. In cell lines with the highest HEY1 induction, NOTCH3 activation was the consequence of JAG1 transcriptional induction. JAG1 modulation by specific siRNA, NOTCH-processing inhibition by either GSI or ectopic NUMB1, and siRNA-mediated HEY1 knockdown all inhibited p53 and p21(WAF1/CIP1) induction. Conversely, forced expression of JAG1, activated NOTCH3, or HEY1 induced p53 and p21(WAF1/CIP1). These results indicate that suppression of EWS-FLI1 reactivates NOTCH signaling in ESFT cells, resulting in p53-dependent cell cycle arrest. Our data link EWS-FLI1 to the NOTCH and p53 pathways and provide a plausible basis both for NOTCH tumor suppressor effects and oncogenesis of cancers that retain wild-type p53.


Subject(s)
Genes, p53 , Oncogene Proteins, Fusion/physiology , Receptors, Notch/physiology , Sarcoma, Ewing/metabolism , Transcription Factors/physiology , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Primers , Fluorescent Antibody Technique , Gene Silencing , Humans , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...