Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Sci Rep ; 11(1): 389, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431989

ABSTRACT

Lentiviral vectors (LV) have emerged as a robust technology for therapeutic gene delivery into human cells as advanced medicinal products. As these products are increasingly commercialized, there are concomitant demands for their characterization to ensure safety, efficacy and consistency. Standards are essential for accurately measuring parameters for such product characterization. A critical parameter is the vector copy number (VCN) which measures the genetic dose of a transgene present in gene-modified cells. Here we describe a set of clonal Jurkat cell lines with defined copy numbers of a reference lentiviral vector integrated into their genomes. Genomic DNA was characterized for copy number, genomic integrity and integration coordinates and showed uniform performance across independent quantitative PCR assays. Stability studies during continuous long-term culture demonstrated sustained renewability of the reference standard source material. DNA from the Jurkat VCN standards would be useful for control of quantitative PCR assays for VCN determination in LV gene-modified cellular products and clinical samples.


Subject(s)
Gene Dosage , Lentivirus/genetics , Transduction, Genetic , Calibration/standards , Gene Transfer Techniques/standards , Genetic Vectors/genetics , Humans , Jurkat Cells , Mutagenesis, Insertional/genetics , Reference Standards , Reproducibility of Results , Transduction, Genetic/methods , Transduction, Genetic/standards , Transfection/methods , Transfection/standards , Validation Studies as Topic , Virus Integration/genetics
2.
Nat Commun ; 10(1): 3718, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31420543

ABSTRACT

High throughput omics approaches provide an unprecedented opportunity for dissecting molecular mechanisms in cancer biology. Here we present deep profiling of whole proteome, phosphoproteome and transcriptome in two high-grade glioma (HGG) mouse models driven by mutated RTK oncogenes, PDGFRA and NTRK1, analyzing 13,860 proteins and 30,431 phosphosites by mass spectrometry. Systems biology approaches identify numerous master regulators, including 41 kinases and 23 transcription factors. Pathway activity computation and mouse survival indicate the NTRK1 mutation induces a higher activation of AKT downstream targets including MYC and JUN, drives a positive feedback loop to up-regulate multiple other RTKs, and confers higher oncogenic potency than the PDGFRA mutation. A mini-gRNA library CRISPR-Cas9 validation screening shows 56% of tested master regulators are important for the viability of NTRK-driven HGG cells, including TFs (Myc and Jun) and metabolic kinases (AMPKa1 and AMPKa2), confirming the validity of the multiomics integrative approaches, and providing novel tumor vulnerabilities.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Profiling , Glioma/genetics , Proteomics , AMP-Activated Protein Kinases/metabolism , Animals , Brain Neoplasms/metabolism , Disease Models, Animal , Feedback, Physiological , Glioma/metabolism , Mice , Mutation , Oncogene Protein p65(gag-jun)/metabolism , Phosphopeptides/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, trkA/genetics , Signal Transduction , Systems Biology , Up-Regulation
3.
Cancer Cell ; 35(1): 140-155.e7, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30595505

ABSTRACT

Diffuse intrinsic pontine gliomas (DIPGs) are incurable childhood brainstem tumors with frequent histone H3 K27M mutations and recurrent alterations in PDGFRA and TP53. We generated genetically engineered inducible mice and showed that H3.3 K27M enhanced neural stem cell self-renewal while preserving regional identity. Neonatal induction of H3.3 K27M cooperated with activating platelet-derived growth factor receptor α (PDGFRα) mutant and Trp53 loss to accelerate development of diffuse brainstem gliomas that recapitulated human DIPG gene expression signatures and showed global changes in H3K27 posttranslational modifications, but relatively restricted gene expression changes. Genes upregulated in H3.3 K27M tumors were enriched for those associated with neural development where H3K27me3 loss released the poised state of apparently bivalent promoters, whereas downregulated genes were enriched for those encoding homeodomain transcription factors.


Subject(s)
Brain Stem Neoplasms/genetics , Gene Expression Profiling/methods , Glioma/genetics , Histones/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Self Renewal , Cells, Cultured , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Mice , Mutation , Neural Stem Cells/cytology , Rhombencephalon/pathology , Sequence Analysis, RNA/methods
4.
Nat Genet ; 46(5): 444-450, 2014 May.
Article in English | MEDLINE | ID: mdl-24705251

ABSTRACT

Pediatric high-grade glioma (HGG) is a devastating disease with a less than 20% survival rate 2 years after diagnosis. We analyzed 127 pediatric HGGs, including diffuse intrinsic pontine gliomas (DIPGs) and non-brainstem HGGs (NBS-HGGs), by whole-genome, whole-exome and/or transcriptome sequencing. We identified recurrent somatic mutations in ACVR1 exclusively in DIPGs (32%), in addition to previously reported frequent somatic mutations in histone H3 genes, TP53 and ATRX, in both DIPGs and NBS-HGGs. Structural variants generating fusion genes were found in 47% of DIPGs and NBS-HGGs, with recurrent fusions involving the neurotrophin receptor genes NTRK1, NTRK2 and NTRK3 in 40% of NBS-HGGs in infants. Mutations targeting receptor tyrosine kinase-RAS-PI3K signaling, histone modification or chromatin remodeling, and cell cycle regulation were found in 68%, 73% and 59% of pediatric HGGs, respectively, including in DIPGs and NBS-HGGs. This comprehensive analysis provides insights into the unique and shared pathways driving pediatric HGG within and outside the brainstem.


Subject(s)
Activin Receptors, Type I/genetics , Brain Stem Neoplasms/genetics , Glioma/genetics , Signal Transduction/genetics , Animals , Child , Cohort Studies , Computational Biology , Gene Expression Profiling , Gene Fusion/genetics , Humans , Immunoblotting , Immunohistochemistry , Microarray Analysis , Receptor, trkA/genetics , Receptor, trkB/genetics , Receptor, trkC/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Statistics, Nonparametric , Zebrafish
5.
Cancer Res ; 73(20): 6219-29, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23970477

ABSTRACT

The outcome for children with high-grade gliomas (HGG) remains dismal, with a 2-year survival rate of only 10% to 30%. Diffuse intrinsic pontine glioma (DIPG) comprise a subset of HGG that arise in the brainstem almost exclusively in children. Genome-wide analyses of copy number imbalances previously showed that platelet-derived growth factor receptor α (PDGFRA) is the most frequent target of focal amplification in pediatric HGGs, including DIPGs. To determine whether PDGFRA is also targeted by more subtle mutations missed by copy number analysis, we sequenced all PDGFRA coding exons from a cohort of pediatric HGGs. Somatic-activating mutations were identified in 14.4% (13 of 90) of nonbrainstem pediatric HGGs and 4.7% (2 of 43) of DIPGs, including missense mutations and in-frame deletions and insertions not previously described. Forty percent of tumors with mutation showed concurrent amplification, whereas 60% carried heterozygous mutations. Six different mutations impacting different domains all resulted in ligand-independent receptor activation that was blocked by small molecule inhibitors of PDGFR. Expression of mutants in p53-null primary mouse astrocytes conferred a proliferative advantage in vitro and generated HGGs in vivo with complete penetrance when implanted into brain. The gene expression signatures of these murine HGGs reflected the spectrum of human diffuse HGGs. PDGFRA intragenic deletion of exons 8 and 9 were previously shown in adult HGG, but were not detected in 83 nonbrainstem pediatric HGG and 57 DIPGs. Thus, a distinct spectrum of mutations confers constitutive receptor activation and oncogenic activity to PDGFRα in childhood HGG.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Mutation , Receptor, Platelet-Derived Growth Factor alpha/genetics , Adolescent , Animals , Brain Neoplasms/pathology , Child , Child, Preschool , Gene Expression Profiling , Genome-Wide Association Study , Glioma/pathology , Humans , Mice , Receptor, Platelet-Derived Growth Factor alpha/metabolism
6.
Nat Genet ; 44(3): 251-3, 2012 Jan 29.
Article in English | MEDLINE | ID: mdl-22286216

ABSTRACT

To identify somatic mutations in pediatric diffuse intrinsic pontine glioma (DIPG), we performed whole-genome sequencing of DNA from seven DIPGs and matched germline tissue and targeted sequencing of an additional 43 DIPGs and 36 non-brainstem pediatric glioblastomas (non-BS-PGs). We found that 78% of DIPGs and 22% of non-BS-PGs contained a mutation in H3F3A, encoding histone H3.3, or in the related HIST1H3B, encoding histone H3.1, that caused a p.Lys27Met amino acid substitution in each protein. An additional 14% of non-BS-PGs had somatic mutations in H3F3A causing a p.Gly34Arg alteration.


Subject(s)
Brain Stem Neoplasms/genetics , Glioblastoma/genetics , Histones/genetics , Pons/pathology , Base Sequence , Brain Stem Neoplasms/pathology , Genome, Human/genetics , Humans , Molecular Sequence Data , Mutation, Missense/genetics , Sequence Analysis, DNA
7.
Clin Cancer Res ; 17(24): 7595-604, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22038996

ABSTRACT

PURPOSE: Malignant astrocytomas (MA) are aggressive central nervous system tumors with poor prognosis. Activating mutation of BRAF (BRAF(V600E)) has been reported in a subset of these tumors, especially in children. We have investigated the incidence of BRAF(V600E) in additional pediatric patient cohorts and examined the effects of BRAF blockade in preclinical models of BRAF(V600E) and wild-type BRAF MA. EXPERIMENTAL DESIGN: BRAF(V600E) mutation status was examined in two pediatric MA patient cohorts. For functional studies, BRAF(V600E) MA cell lines were used to investigate the effects of BRAF shRNA knockdown in vitro, and to investigate BRAF pharmacologic inhibition in vitro and in vivo. RESULTS: BRAF(V600E) mutations were identified in 11 and 10% of MAs from two distinct series of tumors (six of 58 cases total). BRAF was expressed in all MA cell lines examined, among which BRAF(V600E) was identified in four instances. Using the BRAF(V600E)-specific inhibitor PLX4720, pharmacologic blockade of BRAF revealed preferential antiproliferative activity against BRAF(V600E) mutant cells in vitro, in contrast to the use of shRNA-mediated knockdown of BRAF, which inhibited cell growth of glioma cell lines regardless of BRAF mutation status. Using orthotopic MA xenografts, we show that PLX4720 treatment decreases tumor growth and increases overall survival in mice-bearing BRAF(V600E) mutant xenografts, while being ineffective, and possibly tumor promoting, against xenografts with wild-type BRAF. CONCLUSIONS: Our results indicate a 10% incidence of activating BRAF(V600E) among pediatric MAs. With regard to implications for therapy, our results support evaluation of BRAF(V600E)-specific inhibitors for treating BRAF(V600E) MA patients.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Adolescent , Amino Acid Substitution , Animals , Astrocytoma/metabolism , Astrocytoma/pathology , Astrocytoma/prevention & control , Base Sequence , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/prevention & control , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Child, Preschool , DNA Mutational Analysis , Female , Humans , Indoles/pharmacology , Infant , Kaplan-Meier Estimate , Mice , Mice, Nude , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , RNA Interference , Sulfonamides/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Young Adult
8.
J Clin Oncol ; 29(30): 3999-4006, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-21931021

ABSTRACT

PURPOSE: Long-term survival for children with diffuse intrinsic pontine glioma (DIPG) is less than 10%, and new therapeutic targets are urgently required. We evaluated a large cohort of DIPGs to identify recurrent genomic abnormalities and gene expression signatures underlying DIPG. PATIENTS AND METHODS: Single-nucleotide polymorphism arrays were used to compare the frequencies of genomic copy number abnormalities in 43 DIPGs and eight low-grade brainstem gliomas with data from adult and pediatric (non-DIPG) glioblastomas, and expression profiles were evaluated using gene expression arrays for 27 DIPGs, six low-grade brainstem gliomas, and 66 nonbrainstem low-grade gliomas. RESULTS: Frequencies of specific large-scale and focal imbalances varied significantly between DIPGs and nonbrainstem pediatric glioblastomas. Focal amplifications of genes within the receptor tyrosine kinase-Ras-phosphoinositide 3-kinase signaling pathway were found in 47% of DIPGs, the most common of which involved PDGFRA and MET. Thirty percent of DIPGs contained focal amplifications of cell-cycle regulatory genes controlling retinoblastoma protein (RB) phosphorylation, and 21% had concurrent amplification of genes from both pathways. Some tumors showed heterogeneity in amplification patterns. DIPGs showed distinct gene expression signatures related to developmental processes compared with nonbrainstem pediatric high-grade gliomas, whereas expression signatures of low-grade brainstem and nonbrainstem gliomas were similar. CONCLUSION: DIPGs comprise a molecularly related but distinct subgroup of pediatric gliomas. Genomic studies suggest that targeted inhibition of receptor tyrosine kinases and RB regulatory proteins may be useful therapies for DIPG.


Subject(s)
Brain Stem Neoplasms/genetics , Cell Cycle Proteins/genetics , Genes, cdc , Receptor Protein-Tyrosine Kinases/genetics , Adult , Brain Stem Neoplasms/enzymology , Brain Stem Neoplasms/pathology , Child , Gene Amplification , Gene Dosage , Gene Expression Profiling , Genome-Wide Association Study , Humans , In Situ Hybridization, Fluorescence , Polymorphism, Single Nucleotide , Retinoblastoma Protein/genetics
9.
J Clin Oncol ; 28(18): 3061-8, 2010 Jun 20.
Article in English | MEDLINE | ID: mdl-20479398

ABSTRACT

PURPOSE: To define copy number alterations and gene expression signatures underlying pediatric high-grade glioma (HGG). PATIENTS AND METHODS: We conducted a high-resolution analysis of genomic imbalances in 78 de novo pediatric HGGs, including seven diffuse intrinsic pontine gliomas, and 10 HGGs arising in children who received cranial irradiation for a previous cancer using single nucleotide polymorphism microarray analysis. Gene expression was analyzed with gene expression microarrays for 53 tumors. Results were compared with publicly available data from adult tumors. RESULTS: Significant differences in copy number alterations distinguish childhood and adult glioblastoma. PDGFRA was the predominant target of focal amplification in childhood HGG, including diffuse intrinsic pontine gliomas, and gene expression analyses supported an important role for deregulated PDGFRalpha signaling in pediatric HGG. No IDH1 hotspot mutations were found in pediatric tumors, highlighting molecular differences with adult secondary glioblastoma. Pediatric and adult glioblastomas were clearly distinguished by frequent gain of chromosome 1q (30% v 9%, respectively) and lower frequency of chromosome 7 gain (13% v 74%, respectively) and 10q loss (35% v 80%, respectively). PDGFRA amplification and 1q gain occurred at significantly higher frequency in irradiation-induced tumors, suggesting that these are initiating events in childhood gliomagenesis. A subset of pediatric HGGs showed minimal copy number changes. CONCLUSION: Integrated molecular profiling showed substantial differences in the molecular features underlying pediatric and adult HGG, indicating that findings in adult tumors cannot be simply extrapolated to younger patients. PDGFRalpha may be a useful target for pediatric HGG, including diffuse pontine gliomas.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Gene Expression Profiling , Glioma/genetics , Polymorphism, Single Nucleotide/genetics , Adolescent , Adult , Brain Neoplasms/pathology , Child , Child, Preschool , Chromosomes, Human, Pair 1/genetics , Cranial Irradiation , Glioma/pathology , Humans , Infant , Oligonucleotide Array Sequence Analysis , Prognosis , Receptor, Platelet-Derived Growth Factor alpha/genetics , Young Adult
10.
J Biol Chem ; 284(6): 3408-17, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19074142

ABSTRACT

Chronic inflammation and inflammatory cytokines have recently been implicated in the development and progression of various types of cancer. In the brain, neuroinflammatory cytokines affect the growth and differentiation of both normal and malignant glial cells, with interleukin 1 (IL-1) shown to be secreted by the majority of glioblastoma cells. Recently, elevated levels of sphingosine kinase 1 (SphK1), but not SphK2, were correlated with a shorter survival prognosis for patients with glioblastoma multiforme. SphK1 is a lipid kinase that produces the pro-growth, anti-apoptotic sphingosine 1-phosphate, which can induce invasion of glioblastoma cells. Here, we show that the expression of IL-1 correlates with the expression of SphK1 in glioblastoma cells, and neutralizing anti-IL-1 antibodies inhibit both the growth and invasion of glioblastoma cells. Furthermore, IL-1 up-regulates SphK1 mRNA levels, protein expression, and activity in both primary human astrocytes and various glioblastoma cell lines; however, it does not affect SphK2 expression. The IL-1-induced SphK1 up-regulation can be blocked by the inhibition of JNK, the overexpression of the dominant-negative c-Jun(TAM67), and the down-regulation of c-Jun expression by small interference RNA. Activation of SphK1 expression by IL-1 occurs on the level of transcription and is mediated via a novel AP-1 element located within the first intron of the sphk1 gene. In summary, our results suggest that SphK1 expression is transcriptionally regulated by IL-1 in glioblastoma cells, and this pathway may be important in regulating survival and invasiveness of glioblastoma cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Interleukin-1/metabolism , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Response Elements , Cell Line, Tumor , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Interleukin-1/genetics , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Neoplasm Invasiveness , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism
11.
Mol Cancer Res ; 6(9): 1469-77, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18819934

ABSTRACT

Glioblastoma multiforme is an invasive primary brain tumor, which evades the current standard treatments. The invasion of glioblastoma cells into healthy brain tissue partly depends on the proteolytic and nonproteolytic activities of the plasminogen activator system proteins, including the urokinase-type plasminogen activator (uPA), plasminogen activator inhibitor 1 (PAI-1), and a receptor for uPA (uPAR). Here we show that sphingosine-1-phosphate (S1P) and the inflammatory mediator interleukin-1 (IL-1) increase the mRNA and protein expression of PAI-1 and uPAR and enhance the invasion of U373 glioblastoma cells. Although IL-1 enhanced the expression of sphingosine kinase 1 (SphK1), the enzyme that produces S1P, down-regulation of SphK1 had no effect on the IL-1-induced uPAR or PAI-1 mRNA expression, suggesting that these actions of IL-1 are independent of S1P production. Indeed, the S1P-induced mRNA expression of uPAR and PAI-1 was blocked by the S1P(2) receptor antagonist JTE013 and by the down-regulation of S1P(2) using siRNA. Accordingly, the inhibition of mitogen-activated protein kinase/extracellular signal-regulated kinase kinase 1/2 and Rho-kinase, two downstream signaling cascades activated by S1P(2), blocked the activation of PAI-1 and uPAR mRNA expression by S1P. More importantly, the attachment of glioblastoma cells was inhibited by the addition of exogenous PAI-1 or siRNA to uPAR, whereas the invasion of glioblastoma cells induced by S1P or IL-1 correlated with their ability to enhance the expression of PAI-1 and uPAR. Collectively, these results indicate that S1P and IL-1 activate distinct pathways leading to the mRNA and protein expression of PAI-1 and uPAR, which are important for glioblastoma invasiveness.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Interleukin-1/pharmacology , Lysophospholipids/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Receptors, Cell Surface/metabolism , Sphingosine/analogs & derivatives , Blotting, Northern , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Adhesion/physiology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Plasminogen Activator Inhibitor 1/chemistry , Plasminogen Activator Inhibitor 1/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sphingosine/pharmacology , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
12.
Blood ; 112(4): 1382-91, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18511810

ABSTRACT

The potent bioactive sphingolipid mediator, sphingosine-1-phosphate (S1P), is produced by 2 sphingosine kinase isoenzymes, SphK1 and SphK2. Expression of SphK1 is up-regulated in cancers, including leukemia, and associated with cancer progression. A screen of sphingosine analogs identified (2R,3S,4E)-N-methyl-5-(4'-pentylphenyl)-2-aminopent-4-ene-1,3-diol, designated SK1-I (BML-258), as a potent, water-soluble, isoenzyme-specific inhibitor of SphK1. In contrast to pan-SphK inhibitors, SK1-I did not inhibit SphK2, PKC, or numerous other protein kinases. SK1-I decreased growth and survival of human leukemia U937 and Jurkat cells, and enhanced apoptosis and cleavage of Bcl-2. Lethality of SK1-I was reversed by caspase inhibitors and by expression of Bcl-2. SK1-I not only decreased S1P levels but concomitantly increased levels of its proapoptotic precursor ceramide. Conversely, S1P protected against SK1-I-induced apoptosis. SK1-I also induced multiple perturbations in activation of signaling and survival-related proteins, including diminished phosphorylation of ERK1/2 and Akt. Expression of constitutively active Akt protected against SK1-I-induced apoptosis. Notably, SK1-I potently induced apoptosis in leukemic blasts isolated from patients with acute myelogenous leukemia but was relatively sparing of normal peripheral blood mononuclear leukocytes. Moreover, SK1-I markedly reduced growth of AML xenograft tumors. Our results suggest that specific inhibitors of SphK1 warrant attention as potential additions to the therapeutic armamentarium in leukemia.


Subject(s)
Leukemia/drug therapy , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Sphingosine/analogs & derivatives , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Enzyme Inhibitors/therapeutic use , Humans , Mice , Mice, SCID , Proto-Oncogene Proteins c-bcl-2/metabolism , Sphingosine/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
FASEB J ; 22(2): 455-65, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17855624

ABSTRACT

Patients with gliomas expressing high levels of epidermal growth factor receptor (EGFR) and plasminogen activator inhibitor-1 (PAI-1) have a shorter overall survival prognosis. Moreover, EGF enhances PAI-1 expression in glioma cells. Although multiple known signaling cascades are activated by EGF in glioma cells, we show for the first time that EGF enhances expression of PAI-1 via sequential activation of c-Src, protein kinase C delta (PKCdelta), and sphingosine kinase 1 (SphK1), the enzyme that produces sphingosine-1-phosphate. EGF induced rapid phosphorylation of c-Src and PKCdelta and concomitant translocation of PKCdelta as well as SphK1 to the plasma membrane. Down-regulation of PKCdelta abolished EGF-induced SphK1 translocation and up-regulation of PAI-1 by EGF; whereas, down-regulation of PKCalpha had no effect on the EGF-induced PAI-1 activation but enhanced its basal expression. Similarly, inhibition of c-Src activity by PP2 blocked both EGF-induced translocation of SphK1 and PKCdelta to the plasma membrane and up-regulation of PAI-1 expression. Furthermore, SphK1 was indispensable for both EGF-induced c-Jun phosphorylation and PAI-1 expression. Collectively, our results provide a functional link between three critical downstream targets of EGF, c-Src, PKCdelta, and SphK1 that have all been implicated in regulating motility and invasion of glioma cells.


Subject(s)
Epidermal Growth Factor/pharmacology , Glioblastoma/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Protein Kinase C-delta/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction/drug effects , Cell Line, Tumor , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glioblastoma/genetics , Humans , NF-kappa B/metabolism , Plasminogen Activator Inhibitor 1/genetics , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Protein Kinase C-delta/genetics , STAT Transcription Factors/metabolism , Transcription Factor AP-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL