Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 18(9): 1565-1576, 2019 09.
Article in English | MEDLINE | ID: mdl-31270152

ABSTRACT

Amplification of the epidermal growth factor receptor gene (EGFR) represents one of the most commonly observed genetic lesions in glioblastoma (GBM); however, therapies targeting this signaling pathway have failed clinically. Here, using human tumors, primary patient-derived xenografts (PDX), and a murine model for GBM, we demonstrate that EGFR inhibition leads to increased invasion of tumor cells. Further, EGFR inhibitor-treated GBM demonstrates altered oxidative stress, with increased lipid peroxidation, and generation of toxic lipid peroxidation products. A tumor cell subpopulation with elevated aldehyde dehydrogenase (ALDH) levels was determined to comprise a significant proportion of the invasive cells observed in EGFR inhibitor-treated GBM. Our analysis of the ALDH1A1 protein in newly diagnosed GBM revealed detectable ALDH1A1 expression in 69% (35/51) of the cases, but in relatively low percentages of tumor cells. Analysis of paired human GBM before and after EGFR inhibitor therapy showed an increase in ALDH1A1 expression in EGFR-amplified tumors (P < 0.05, n = 13 tumor pairs), and in murine GBM ALDH1A1-high clones were more resistant to EGFR inhibition than ALDH1A1-low clones. Our data identify ALDH levels as a biomarker of GBM cells with high invasive potential, altered oxidative stress, and resistance to EGFR inhibition, and reveal a therapeutic target whose inhibition should limit GBM invasion.


Subject(s)
Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Glioblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays/methods , Aldehyde Dehydrogenase 1 Family/metabolism , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dasatinib/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Oxidative Stress/drug effects , Retinal Dehydrogenase/metabolism
2.
Nat Cell Biol ; 20(10): 1203-1214, 2018 10.
Article in English | MEDLINE | ID: mdl-30202050

ABSTRACT

Glioblastoma multiforme (GBMs) are recurrent lethal brain tumours. Recurrent GBMs often exhibit mesenchymal, stem-like phenotypes that could explain their resistance to therapy. Analyses revealed that recurrent GBMs have increased tension and express high levels of glycoproteins that increase the bulkiness of the glycocalyx. Studies showed that a bulky glycocalyx potentiates integrin mechanosignalling and tissue tension and promotes a mesenchymal, stem-like phenotype in GBMs. Gain- and loss-of-function studies implicated integrin mechanosignalling as an inducer of GBM growth, survival, invasion and treatment resistance, and a mesenchymal, stem-like phenotype. Mesenchymal-like GBMs were highly contractile and expressed elevated levels of glycoproteins that expanded their glycocalyx, and they were surrounded by a stiff extracellular matrix that potentiated integrin mechanosignalling. Our findings suggest that there is a dynamic and reciprocal link between integrin mechanosignalling and a bulky glycocalyx, implying a causal link towards a mesenchymal, stem-like phenotype in GBMs. Strategies to ameliorate GBM tissue tension offer a therapeutic approach to reduce mortality due to GBM.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Glycocalyx/metabolism , Integrins/metabolism , Mesenchymal Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Survival/drug effects , Feedback, Physiological/drug effects , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Mesenchymal Stem Cells/drug effects , Mice, Nude , Neoplastic Stem Cells/drug effects , Surface Tension , Temozolomide/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
CNS Oncol ; 2(1): 49-65, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23750318

ABSTRACT

Angiogenesis inhibitors targeting the VEGF signaling pathway have been US FDA approved for various cancers including glioblastoma (GBM), one of the most lethal and angiogenic tumors. This has led to the routine use of the anti-VEGF antibody bevacizumab in recurrent GBM, conveying substantial improvements in radiographic response, progression-free survival and quality of life. Despite these encouraging beneficial effects, patients inevitably develop resistance and frequently fail to demonstrate significantly better overall survival. Unlike chemotherapies, to which tumors exhibit resistance due to genetic mutation of drug targets, emerging evidence suggests that tumors bypass antiangiogenic therapy while VEGF signaling remains inhibited through a variety of mechanisms that are just beginning to be recognized. Because of the indirect nature of resistance to VEGF inhibitors there is promise that strategies combining angiogenesis inhibitors with drugs targeting such evasive resistance pathways will lead to more durable antiangiogenic efficacy and improved patient outcomes. Further identifying and understanding of evasive resistance mechanisms and their clinical importance in GBM relapse is therefore a timely and critical issue.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Glioblastoma/metabolism , Vascular Endothelial Growth Factor A/metabolism , Humans , Signal Transduction/drug effects
4.
Cancer Cell ; 22(1): 21-35, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22789536

ABSTRACT

Inhibition of VEGF signaling leads to a proinvasive phenotype in mouse models of glioblastoma multiforme (GBM) and in a subset of GBM patients treated with bevacizumab. Here, we demonstrate that vascular endothelial growth factor (VEGF) directly and negatively regulates tumor cell invasion through enhanced recruitment of the protein tyrosine phosphatase 1B (PTP1B) to a MET/VEGFR2 heterocomplex, thereby suppressing HGF-dependent MET phosphorylation and tumor cell migration. Consequently, VEGF blockade restores and increases MET activity in GBM cells in a hypoxia-independent manner, while inducing a program reminiscent of epithelial-to-mesenchymal transition highlighted by a T-cadherin to N-cadherin switch and enhanced mesenchymal features. Inhibition of MET in GBM mouse models blocks mesenchymal transition and invasion provoked by VEGF ablation, resulting in substantial survival benefit.


Subject(s)
Brain Neoplasms/pathology , Epithelial-Mesenchymal Transition/physiology , Glioblastoma/pathology , Proto-Oncogene Proteins c-met/metabolism , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Bevacizumab , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Phosphorylation , Proto-Oncogene Proteins c-met/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
5.
Cancer Res ; 69(17): 6889-98, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19690143

ABSTRACT

Activating epidermal growth factor receptor (EGFR) mutations are common in many cancers including glioblastoma. However, clinical responses to EGFR inhibitors are infrequent and short-lived. We show that the Src family kinases (SFK) Fyn and Src are effectors of oncogenic EGFR signaling, enhancing invasion and tumor cell survival in vivo. Expression of a constitutively active EGFR mutant, EGFRvIII, resulted in activating phosphorylation and physical association with Src and Fyn, promoting tumor growth and motility. Gene silencing of Fyn and Src limited EGFR- and EGFRvIII-dependent tumor cell motility. The SFK inhibitor dasatinib inhibited invasion, promoted tumor regression, and induced apoptosis in vivo, significantly prolonging survival of an orthotopic glioblastoma model expressing endogenous EGFRvIII. Dasatinib enhanced the efficacy of an anti-EGFR monoclonal antibody (mAb 806) in vivo, further limiting tumor growth and extending survival. Examination of a large cohort of clinical samples showed frequent coactivation of EGFR and SFKs in glioblastoma patients. These results establish a mechanism linking EGFR signaling with Fyn and Src activation to promote tumor progression and invasion in vivo and provide rationale for combined anti-EGFR and anti-SFK targeted therapies.


Subject(s)
Central Nervous System Neoplasms/metabolism , ErbB Receptors/metabolism , Glioblastoma/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Signal Transduction , src-Family Kinases/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Central Nervous System Neoplasms/pathology , Dasatinib , Drug Synergism , Enzyme Activation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Gene Silencing , Glioblastoma/pathology , Humans , Mice , Mutation , Neoplasm Invasiveness , Neoplasm Transplantation , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology
6.
Cancer Res ; 68(24): 10034-9, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19074867

ABSTRACT

In this study, we investigated the precursor and active forms of a p53 small-molecule inhibitor for their effects on temozolomide (TMZ) antitumor activity against glioblastoma (GBM), using both in vitro and in vivo experimental approaches. Results from in vitro cell viability analysis showed that the cytotoxic activity of TMZ was substantially increased when p53 wild-type (p53(wt)) GBMs were cotreated with the active form of p53 inhibitor, and this heightened cytotoxic response was accompanied by increased poly(ADP-ribose) polymerase cleavage as well as elevated cellular phospho-H2AX. Analysis of the same series of GBMs, as intracranial xenografts in athymic mice, and administering corresponding p53 inhibitor precursor, which is converted to the active compound in vivo, yielded results consistent with the in vitro analyses: TMZ + p53 inhibitor precursor cotreatment of three distinct p53(wt) GBM xenografts resulted in significant enhancement of TMZ antitumor effect relative to treatment with TMZ alone, as indicated by serial bioluminescence monitoring as well as survival analysis (P < 0.001 for cotreatment survival benefit in each case). Mice receiving intracranial injection with p53(null) GBM showed similar survival benefit from TMZ treatment regardless of the presence or absence of p53 inhibitor precursor. In total, our results indicate that the p53 active and precursor inhibitor pair enhances TMZ cytotoxicity in vitro and in vivo, respectively, and do so in a p53-dependent manner.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Benzothiazoles , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Dacarbazine/administration & dosage , Dacarbazine/pharmacology , Drug Synergism , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Poly(ADP-ribose) Polymerases/metabolism , Temozolomide , Toluene/analogs & derivatives , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
7.
Cancer Cell ; 13(3): 206-20, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18328425

ABSTRACT

Development of hypoxic regions is an indicator of poor prognosis in many tumors. Here, we demonstrate that HIF1alpha, the direct effector of hypoxia, partly through increases in SDF1alpha, induces recruitment of bone marrow-derived CD45+ myeloid cells containing Tie2+, VEGFR1+, CD11b+, and F4/80+ subpopulations, as well as endothelial and pericyte progenitor cells to promote neovascularization in glioblastoma. MMP-9 activity of bone marrow-derived CD45+ cells is essential and sufficient to initiate angiogenesis by increasing VEGF bioavailability. In the absence of HIF1alpha, SDF1alpha levels decrease, and fewer BM-derived cells are recruited to the tumors, decreasing MMP-9 and mobilization of VEGF. VEGF also directly regulates tumor cell invasiveness. When VEGF activity is impaired, tumor cells invade deep into the brain in the perivascular compartment.


Subject(s)
Bone Marrow Cells/enzymology , Brain Neoplasms/blood supply , Glioblastoma/blood supply , Glioblastoma/enzymology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Matrix Metalloproteinase 9/metabolism , Neovascularization, Pathologic/enzymology , Animals , Antigens, Differentiation/metabolism , Benzylamines , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cell Hypoxia , Cell Line , Cell Movement , Chemokine CXCL12/metabolism , Cyclams , Endothelial Cells/enzymology , Glioblastoma/pathology , Heterocyclic Compounds/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Leukocyte Common Antigens/metabolism , Matrix Metalloproteinase 9/deficiency , Matrix Metalloproteinase 9/genetics , Mice , Mice, Knockout , Monocytes/enzymology , Neoplasm Invasiveness , Neovascularization, Pathologic/pathology , Pericytes/enzymology , Receptor, TIE-2/metabolism , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Signal Transduction , Transduction, Genetic , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
8.
Cancer Res ; 66(16): 7864-9, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16912159

ABSTRACT

The epidermal growth factor receptor (EGFR) is commonly amplified, overexpressed, and mutated in glioblastoma, making it a compelling molecular target for therapy. We have recently shown that coexpression of EGFRvIII and PTEN protein by glioblastoma cells is strongly associated with clinical response to EGFR kinase inhibitor therapy. PTEN loss, by dissociating inhibition of the EGFR from downstream phosphatidylinositol 3-kinase (PI3K) pathway inhibition, seems to act as a resistance factor. Because 40% to 50% of glioblastomas are PTEN deficient, a critical challenge is to identify strategies that promote responsiveness to EGFR kinase inhibitors in patients whose tumors lack PTEN. Here, we show that the mammalian target of rapamycin (mTOR) inhibitor rapamycin enhances the sensitivity of PTEN-deficient tumor cells to the EGFR kinase inhibitor erlotinib. In two isogenic model systems (U87MG glioblastoma cells expressing EGFR, EGFRvIII, and PTEN in relevant combinations, and SF295 glioblastoma cells in which PTEN protein expression has been stably restored), we show that combined EGFR/mTOR kinase inhibition inhibits tumor cell growth and has an additive effect on inhibiting downstream PI3K pathway signaling. We also show that combination therapy provides added benefit in promoting cell death in PTEN-deficient tumor cells. These studies provide strong rationale for combined mTOR/EGFR kinase inhibitor therapy in glioblastoma patients, particularly those with PTEN-deficient tumors.


Subject(s)
ErbB Receptors/genetics , Glioblastoma/pathology , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinases/physiology , Cell Division/drug effects , Cell Line, Tumor , Enzyme Activation , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Glioblastoma/genetics , Humans , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Quinazolines/pharmacology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Transfection
9.
N Engl J Med ; 353(19): 2012-24, 2005 Nov 10.
Article in English | MEDLINE | ID: mdl-16282176

ABSTRACT

BACKGROUND: The epidermal growth factor receptor (EGFR) is frequently amplified, overexpressed, or mutated in glioblastomas, but only 10 to 20 percent of patients have a response to EGFR kinase inhibitors. The mechanism of responsiveness of glioblastomas to these inhibitors is unknown. METHODS: We sequenced kinase domains in the EGFR and human EGFR type 2 (Her2/neu) genes and analyzed the expression of EGFR, EGFR deletion mutant variant III (EGFRvIII), and the tumor-suppressor protein PTEN in recurrent malignant gliomas from patients who had received EGFR kinase inhibitors. We determined the molecular correlates of clinical response, validated them in an independent data set, and identified effects of the molecular abnormalities in vitro. RESULTS: Of 49 patients with recurrent malignant glioma who were treated with EGFR kinase inhibitors, 9 had tumor shrinkage of at least 25 percent. Pretreatment tissue was available for molecular analysis from 26 patients, 7 of whom had had a response and 19 of whom had rapid progression during therapy. No mutations in EGFR or Her2/neu kinase domains were detected in the tumors. Coexpression of EGFRvIII and PTEN was significantly associated with a clinical response (P<0.001; odds ratio, 51; 95 percent confidence interval, 4 to 669). These findings were validated in 33 patients who received similar treatment for glioblastoma at a different institution (P=0.001; odds ratio, 40; 95 percent confidence interval, 3 to 468). In vitro, coexpression of EGFRvIII and PTEN sensitized glioblastoma cells to erlotinib. CONCLUSIONS: Coexpression of EGFRvIII and PTEN by glioblastoma cells is associated with responsiveness to EGFR kinase inhibitors.


Subject(s)
ErbB Receptors/genetics , Glioblastoma/genetics , PTEN Phosphohydrolase/metabolism , Protein Kinase Inhibitors/therapeutic use , Adult , Aged , Antineoplastic Agents/therapeutic use , DNA, Neoplasm/analysis , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Female , Gefitinib , Gene Amplification , Gene Deletion , Gene Expression , Genes, erbB-1 , Genes, erbB-2 , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Male , Middle Aged , Mutation , Oligodendroglioma/drug therapy , Oligodendroglioma/genetics , Oligodendroglioma/metabolism , PTEN Phosphohydrolase/genetics , Polymerase Chain Reaction , Quinazolines/therapeutic use , Sequence Analysis, DNA , Signal Transduction
10.
J Biol Chem ; 280(29): 26953-64, 2005 Jul 22.
Article in English | MEDLINE | ID: mdl-15908427

ABSTRACT

Glioblastoma is the most common malignant brain tumor of adults and one of the most lethal cancers. The secreted growth factor pleiotrophin (PTN) promotes glioblastoma migration and proliferation, initiating its oncogenic activities through two cell surface receptors, the protein tyrosine phosphatase receptor zeta (PTPRZ1) and the anaplastic lymphoma kinase (ALK), respectively. Here, we report on the presence and purification of two naturally occurring forms of PTN (18 and 15 kDa) that differentially promote glioblastoma migration and proliferation. Using a panel of glioblastoma cell lines, including low passage patient-derived cultures, we demonstrate that PTN15 promotes glioblastoma proliferation in an ALK-dependent fashion, whereas immobilized PTN18 promotes haptotactic migration of glioblastoma cells in a PTPRZ1-dependent fashion. Mass spectrometric analysis indicated that PTN15 differs from PTN18 by processing of 12 C-terminal amino acids. To demonstrate clinical relevance, we show that PTN15, PTN18, and PTPRZ1 are significantly overexpressed in glioblastoma relative to normal brain at both mRNA and protein levels using microarray, Western blot, and tissue microarray analyses on human tumors. These results indicate that the PTN18-PTPRZ1 and the PTN15-ALK signaling pathways represent potentially important therapeutic targets for glioblastoma invasion and growth.


Subject(s)
Carrier Proteins/physiology , Cytokines/physiology , Glioblastoma/pathology , Adult , Anaplastic Lymphoma Kinase , Carrier Proteins/analysis , Carrier Proteins/metabolism , Cell Movement , Cell Proliferation , Cytokines/analysis , Cytokines/metabolism , Glioblastoma/chemistry , Humans , Neoplasm Invasiveness/pathology , Neoplasm Proteins/analysis , Nerve Growth Factors/physiology , Protein Processing, Post-Translational , Protein Tyrosine Phosphatases/analysis , Protein Tyrosine Phosphatases/physiology , Protein-Tyrosine Kinases , RNA, Neoplasm/analysis , Receptor Protein-Tyrosine Kinases , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Tumor Cells, Cultured
11.
Lab Invest ; 84(1): 8-20, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14631378

ABSTRACT

Local invasiveness is a characteristic feature of glioblastoma that makes surgical resection nearly impossible and accounts in large part for its poor prognosis. To identify mechanisms underlying glioblastoma invasion and motility, we used Transwell invasion chambers to select for a more potently invasive subpopulation of U87MG human glioblastoma cells. The stable population of tumor cells (U87-C1) obtained through this in vitro selection process were three times more invasive than parental U87MG cells and demonstrated faster monolayer wound healing and enhanced radial motility from cell spheroids. This enhanced invasiveness was associated with an 80% increase in matrix metalloproteinase 2 (MMP-2) activation. No differences in expression levels of pro-MMP-2, membrane-type matrix metalloproteinase I (MT1-MMP), or integrin alphavbeta3 (mediators of MMP-2 activation) were detected. However, U87-C1 cells exhibited two-fold elevation of tissue inhibitor of metalloproteinases (TIMP)-2 mRNA and protein relative to parental cells. Exogenous addition of comparable levels of purified TIMP-2 to parental U87MG cells increased MMP-2 activation and invasion. Similarly, U87MG cells engineered to overexpress TIMP-2 at the same levels as U87-C1 cells also demonstrated increased MMP-2 activation, indicating that an increase in physiological levels of TIMP-2 can promote MMP-2 activation and invasion in glioblastoma cells. However, exogenous administration or recombinant overexpression of higher amounts of TIMP-2 in U87MG cells resulted in inhibition of MMP-2 activation. These results demonstrate that the complex balance between TIMP-2 and MMP-2 is a critical determinant of glioblastoma invasion, and indicate that increasing TIMP-2 in glioblastoma patients may potentially cause adverse effects, particularly in tumors containing high levels of MT1-MMP and MMP-2.


Subject(s)
Glioblastoma/metabolism , Matrix Metalloproteinase 2/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/metabolism , Dose-Response Relationship, Drug , Glioblastoma/drug therapy , Humans , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/genetics , Neoplasm Invasiveness , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Transfection , Up-Regulation
12.
Oncogene ; 22(15): 2361-73, 2003 Apr 17.
Article in English | MEDLINE | ID: mdl-12700671

ABSTRACT

Epidermal growth factor receptor (EGFR) overexpression occurs in nearly 50% of cases of glioblastoma (GBM), but its clinical and biological implications are not well understood. We have used Affymetrix high-density oligonucleotide arrays to demonstrate that EGFR-overexpressing GBMs (EGFR+) have a distinct global gene transcriptional profile. We show that the expression of 90 genes can distinguish EGFR+ from EGFR nonexpressing (EGFR-) GBMs, including a number of genes known to act as growth/survival factors for GBMs. We have also uncovered two additional novel molecular subtypes of GBMs, one of which is characterized by coordinate upregulation of contiguous genes on chromosome 12q13-15 and expression of both astrocytic and oligodendroglial genes. These results define distinct molecular subtypes of GBMs that may be important in disease stratification, and in the discovery and assessment of GBM treatment strategies.


Subject(s)
Brain Neoplasms/classification , Gene Expression Profiling , Glioblastoma/classification , Oligonucleotide Array Sequence Analysis , Astrocytes/metabolism , Astrocytes/pathology , Biopsy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Differentiation , Cell Lineage , Chromosomes, Human, Pair 12/genetics , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oligodendroglia/metabolism , Oligodendroglia/pathology , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Subtraction Technique , Transcription, Genetic
13.
Mol Genet Metab ; 76(2): 100-10, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12083807

ABSTRACT

Arginase I (AI) has a critical function in mammalian liver as the final enzyme in the urea cycle responsible for the disposal of ammonia from protein catabolism. AI is also expressed in various extrahepatic tissues and may play a role in regulating arginine levels and in providing ornithine for biosynthetic reactions that generate various critical intermediary metabolites such as glutamate, glutamine, GABA, agmatine, polyamines, creatine, proline, and nitric oxide. AI is expressed in red blood cells (RBCs) only in humans and certain higher primates. Macaca fascicularis has been identified as an evolutionary transition species in which RBC-AI expression is co-dominantly regulated. The M. fascicularis AI gene was analyzed to understand AI expression in erythrocytes. Erythroid progenitor cells [nucleated red blood cells (nRBCs)] isolated from cord blood were utilized to demonstrate AI expression by immunocytochemical staining using anti-AI antibody. Introduction of EGFP reporter vectors into nRBC showed that the proximal 1.2 kbp upstream of the AI gene is sufficient for AI expression. Expression of a second arginase isoform, AII, in nRBCs was discovered by cDNA profiling. This contrasts with mature fetal or adult RBCs which contain only the AI protein. In addition, an alternatively spliced AI (AI(')) variant was observed from erythroid mRNA analysis with an alternative splice acceptor site located within intron 2, causing the insertion of eight additional amino acids yet retaining significant enzymatic activity.


Subject(s)
Arginase/genetics , Erythrocytes/enzymology , Liver/enzymology , Alternative Splicing , Animals , Arginase/blood , Base Sequence , Cells, Cultured , DNA, Complementary/genetics , Gene Expression , Genes, Reporter , Humans , Macaca fascicularis/genetics , Macaca fascicularis/metabolism , Molecular Sequence Data , RNA, Messenger/blood , RNA, Messenger/genetics , Sequence Homology, Nucleic Acid , Species Specificity , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...