Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Oncogene ; 34(31): 4078-88, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-25347738

ABSTRACT

In glioblastoma (GBM), the EGF receptor (EGFR) and Src family kinases (SFKs) contribute to an aggressive phenotype. EGFR may be targeted therapeutically; however, resistance to EGFR-targeting drugs such as Erlotinib and Gefitinib develops quickly. In many GBMs, a truncated form of the EGFR (EGFRvIII) is expressed. Although EGFRvIII is constitutively active and promotes cancer progression, its activity is attenuated compared with EGF-ligated wild-type EGFR, suggesting that EGFRvIII may function together with other signaling receptors in cancer cells to induce an aggressive phenotype. In this study, we demonstrate that in EGFRvIII-expressing GBM cells, the urokinase receptor (uPAR) functions as a major activator of SFKs, controlling phosphorylation of downstream targets, such as p130Cas and Tyr-845 in the EGFR in vitro and in vivo. When EGFRvIII expression in GBM cells was neutralized, either genetically or by treating the cells with Gefitinib, paradoxically, the cells demonstrated increased cell migration. The increase in cell migration was explained by a compensatory increase in expression of urokinase-type plasminogen activator, which activates uPAR-dependent cell signaling. GBM cells that were selected for their ability to grow in vivo in the absence of EGFRvIII also demonstrated increased cell migration, due to activation of the uPAR signaling system. The increase in GBM cell migration, induced by genetic or pharmacologic targeting of the EGFR, was blocked by Dasatinib, highlighting the central role of SFKs in uPAR-promoted cell migration. These results suggest that compensatory activation of uPAR-dependent cell signaling, in GBM cells treated with targeted therapeutics, may adversely affect the course of the disease by promoting cell migration, which may be associated with tumor progression.


Subject(s)
Brain Neoplasms/pathology , Cell Movement/drug effects , ErbB Receptors/antagonists & inhibitors , Glioblastoma/pathology , Quinazolines/pharmacology , Receptors, Urokinase Plasminogen Activator/metabolism , Animals , Brain Neoplasms/genetics , Cell Movement/genetics , ErbB Receptors/genetics , Gefitinib , Glioblastoma/genetics , Humans , Mice , Mice, Nude , Phosphorylation/drug effects , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Tumor Cells, Cultured , src-Family Kinases/metabolism
2.
Oncogene ; 34(10): 1270-9, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-24662834

ABSTRACT

Intratumoral heterogeneity (ITH) represents an obstacle for cancer diagnosis and treatment, but little is known about its functional role in cancer progression. The A Desintegrin And Metalloproteinase 23 (ADAM23) gene is epigenetically silenced in different types of tumors, and silencing is often associated with advanced disease and metastasis. Here, we show that invasive breast tumors exhibit significant ADAM23-ITH and that this heterogeneity is critical for tumor growth and metastasis. We demonstrate that while loss of ADAM23 expression enhances invasion, it causes a severe proliferative deficiency and is not itself sufficient to trigger metastasis. Rather, we observed that, in ADAM23-heterotypic environments, ADAM23-negative cells promote tumor growth and metastasis by enhancing the proliferation and invasion of adjacent A23-positive cells through the production of LGI4 (Leucine-rich Glioma Inactivated 4) and nitric oxide (NO). Ablation of LGI4 and NO in A23-negative cells significantly attenuates A23-positive cell proliferation and invasion. Our work denotes a driving role of ADAM23-ITH during disease progression, shifting the malignant phenotype from the cellular to the tissue level. Our findings also provide insights for therapeutic intervention, enforcing the need to ascertain ITH to improve cancer diagnosis and therapy.


Subject(s)
ADAM Proteins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Extracellular Matrix Proteins/metabolism , Nitric Oxide/metabolism , ADAM Proteins/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Epigenesis, Genetic , Extracellular Matrix Proteins/genetics , Female , Gene Silencing , Humans , Neoplasm Metastasis , Nerve Tissue Proteins , Tumor Burden , Tumor Microenvironment
4.
Oncogene ; 33(19): 2504-12, 2014 May 08.
Article in English | MEDLINE | ID: mdl-23728337

ABSTRACT

Glioblastomas (GBMs), the most common and malignant brain tumors, are highly resistant to current therapies. The failure of targeted therapies against aberrantly activated oncogenic signaling, such as that of the EGFR-PI3K/Akt pathway, underscores the urgent need to understand alternative downstream pathways and to identify new molecular targets for the development of more effective treatments for gliomas. Here, we report that EGFRvIII (ΔEGFR/de2-7EGFR), a constitutively active EGFR mutant that is frequently co-overexpressed with EGFR in clinical GBM tumors, promotes glioma growth and invasion through protein kinase A (PKA)-dependent phosphorylation of Dock180, a bipartite guanine nucleotide exchange factor (GEF) for Rac1. We demonstrate that EGFRvIII induces serine phosphorylation of Dock180, stimulates Rac1 activation and glioma cell migration. Treatments of glioma cells using the PKA inhibitors H-89 and KT5720, overexpression of a PKA inhibitor (PKI), and in vitro PKA kinase assays show that EGFRvIII induction of serine phosphorylation of Dock180 is PKA-dependent. Significantly, PKA induces phosphorylation of Dock180 at amino acid residue S1250 that resides within its Rac1-activating DHR-2 domain. Expression of the Dock180(S1250L) mutant, but not wild type Dock180(WT), protein in EGFRvIII-expressing glioma cells inhibited receptor-stimulated cell proliferation, survival, migration in vitro and glioma tumor growth and invasion in vivo. Together, our findings describe a novel mechanism by which EGFRvIII drives glioma tumorigenesis and invasion through PKA-dependent phosphorylation of Dock180, thereby suggesting that targeting EGFRvIII-PKA-Dock180-Rac1 signaling axis could provide a novel pathway to develop potential therapeutic strategies for malignant gliomas.


Subject(s)
ErbB Receptors/metabolism , Glioma/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Glioma/pathology , HEK293 Cells , Heterografts , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Nude , Phosphorylation , Serine/metabolism
5.
Curr Cancer Drug Targets ; 12(3): 197-209, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22268382

ABSTRACT

Glioblastoma (glioblastoma multiforme; GBM; WHO Grade IV) accounts for the majority of primary malignant brain tumors in adults. Amplification and mutation of the epidermal growth factor receptor (EGFR) gene represent signature genetic abnormalities encountered in GBM. A range of potential therapies that target EGFR or its mutant constitutively active form, ΔEGFR, including tyrosine kinase inhibitors (TKIs), monoclonal antibodies, vaccines, and RNA-based agents, are currently in development or in clinical trials for the treatment of GBM. Data from experimental studies evaluating these therapies have been very promising; however, their efficacy in the clinic has so far been limited by both upfront and acquired drug resistance. This review discusses the current status of anti-EGFR agents and the recurrent problem of resistance to these agents that strongly indicates that a multiple target approach will provide a more favorable future for these types of targeted therapies in GBM.


Subject(s)
Brain Neoplasms/drug therapy , Drug Delivery Systems/methods , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Drug Resistance, Neoplasm/physiology , ErbB Receptors/genetics , Glioblastoma/genetics , Humans , Treatment Outcome
6.
Oncogene ; 31(36): 4054-66, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22139077

ABSTRACT

Sustaining a high growth rate requires tumors to exploit resources in their microenvironment. One example of this is the extensive angiogenesis that is a typical feature of high-grade gliomas. Here, we show that expression of the constitutively active mutant epidermal growth factor receptor, ΔEGFR (EGFRvIII, EGFR*, de2-7EGFR) is associated with significantly higher expression levels of the pro-angiogenic factor interleukin (IL)-8 in human glioma specimens and glioma stem cells. Furthermore, the ectopic expression of ΔEGFR in different glioma cell lines caused up to 60-fold increases in the secretion of IL-8. Xenografts of these cells exhibit increased neovascularization, which is not elicited by cells overexpressing wild-type (wt)EGFR or ΔEGFR with an additional kinase domain mutation. Analysis of the regulation of IL-8 by site-directed mutagenesis of its promoter showed that ΔEGFR regulates its expression through the transcription factors nuclear factor (NF)-κB, activator protein 1 (AP-1) and CCAAT/enhancer binding protein (C/EBP). Glioma cells overexpressing ΔEGFR showed constitutive activation and DNA binding of NF-κB, overexpression of c-Jun and activation of its upstream kinase c-Jun N-terminal kinase (JNK) and overexpression of C/EBPß. Selective pharmacological or genetic targeting of the NF-κB or AP-1 pathways efficiently blocked promoter activity and secretion of IL-8. Moreover, RNA interference-mediated knock-down of either IL-8 or the NF-κB subunit p65, in ΔEGFR-expressing cells attenuated their ability to form tumors and to induce angiogenesis when injected subcutaneously into nude mice. On the contrary, the overexpression of IL-8 in glioma cells lacking ΔEGFR potently enhanced their tumorigenicity and produced highly vascularized tumors, suggesting the importance of this cytokine and its transcription regulators in promoting glioma angiogenesis and tumor growth.


Subject(s)
Glioblastoma/blood supply , Interleukin-8/metabolism , NF-kappa B/metabolism , Neovascularization, Pathologic/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , ErbB Receptors , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Human Umbilical Vein Endothelial Cells/physiology , Humans , Interleukin-8/genetics , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Mice , Mice, Nude , NF-kappa B/antagonists & inhibitors , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Nitriles/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , Response Elements , Sulfones/pharmacology , Transcription Factor AP-1/metabolism , Transcriptional Activation , Tumor Burden , ras Proteins/metabolism
7.
J Biol Chem ; 276(49): 46313-8, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11514572

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive type of glioma and GBMs frequently contain amplifications or mutations of the EGFR gene. The most common mutation results in a truncated receptor tyrosine kinase known as Delta EGFR that signals constitutively and promotes GBM growth. Here, we report that the 45-kDa variant of the protein tyrosine phosphatase TCPTP (TC45) can recognize Delta EGFR as a cellular substrate. TC45 dephosphorylated Delta EGFR in U87MG glioblastoma cells and inhibited mitogen-activated protein kinase ERK2 and phosphatidylinositol 3-kinase signaling. In contrast, the substrate-trapping TC45-D182A mutant, which is capable of forming stable complexes with TC45 substrates, suppressed the activation of ERK2 but not phosphatidylinositol 3-kinase. TC45 inhibited the proliferation and anchorage-independent growth of Delta EGFR cells but TC45-D182A only inhibited cellular proliferation. Notably, neither TC45 nor TC45-D182A inhibited the proliferation of U87MG cells that did not express Delta EGFR. Delta EGFR activity was necessary for the activation of ERK2, and pharmacological inhibition of ERK2 inhibited the proliferation of Delta EGFR-expressing U87MG cells. Expression of either TC45 or TC45-D182A also suppressed the growth of Delta EGFR-expressing U87MG cells in vivo and prolonged the survival of mice implanted intracerebrally with these tumor cells. These results indicate that TC45 can inhibit the Delta EGFR-mediated activation of ERK2 and suppress the tumorigenicity of Delta EGFR-expressing glioblastoma cells in vivo.


Subject(s)
Brain Neoplasms/pathology , ErbB Receptors/genetics , Glioblastoma/pathology , Mutation , Protein Tyrosine Phosphatases/physiology , Signal Transduction/physiology , Animals , Base Sequence , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Cell Division , Cell Line , DNA Primers , ErbB Receptors/metabolism , Female , Flow Cytometry , Glioblastoma/enzymology , Glioblastoma/genetics , Humans , Mice , Mice, Inbred BALB C , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Survival Analysis
8.
Proc Natl Acad Sci U S A ; 98(17): 9936-41, 2001 Aug 14.
Article in English | MEDLINE | ID: mdl-11481434

ABSTRACT

Patients infected with Trypanosoma cruzi may remain asymptomatic for decades and show signs of neuroregeneration in the peripheral nervous system (PNS). In the absence of such neuroregeneration, patients may die in part by extensive neuronal destruction in the gastrointestinal tract. Thus, T. cruzi may, despite their invasion of the PNS, directly prevent cell death to keep nerve destruction in check. Indeed, T. cruzi invasion of Schwann cells, their prime target in PNS, suppressed host-cell apoptosis caused by growth-factor deprivation. The trans-sialidase (TS) of T. cruzi and the Cys-rich domain of TS reproduced the antiapoptotic activity of the parasites at doses (> or =3.0 nM) comparable or lower than those of bona fide mammalian growth factors. This effect was blocked by LY294002, an inhibitor of phosphatidylinositol 3-kinase (PI3K). TS also activated Akt, a downstream effector of PI3K. Ectopic expression of TS in an unrelated parasite, Leishmania major, turned those parasites into activators of Akt in Schwann cells. In contrast, the Cys-rich domain of TS did not block apoptosis in Schwann cells overexpressing dominant-negative Akt or constitutively active PTEN, a negative regulator of PI3K/Akt signaling. The results demonstrate that T. cruzi, through its TS, triggers the survival of host Schwann cells via the PI3K/Akt pathway, suggesting a role for PI3K/Akt in the pathogenesis of Chagas' disease.


Subject(s)
Glycoproteins/pharmacology , Neuraminidase/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Protozoan Proteins/pharmacology , Schwann Cells/drug effects , Signal Transduction/drug effects , Trypanosoma cruzi/enzymology , Tumor Suppressor Proteins , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Chagas Disease/enzymology , Chromones/pharmacology , Culture Media, Serum-Free/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Glycoproteins/genetics , Glycoproteins/physiology , Humans , Leishmania major/enzymology , Molecular Sequence Data , Morpholines/pharmacology , Neuraminidase/genetics , Neuraminidase/physiology , PTEN Phosphohydrolase , Phosphoinositide-3 Kinase Inhibitors , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/physiology , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Recombinant Fusion Proteins/physiology , Schwann Cells/cytology , Schwann Cells/parasitology , Transfection , Trypanosoma cruzi/pathogenicity
10.
Int J Oncol ; 18(5): 1023-6, 2001 May.
Article in English | MEDLINE | ID: mdl-11295051

ABSTRACT

The sensitivity of non-isotopic PCR-SSCP was compared to direct sequencing of PTEN exons. DNA from leukocytes derived from healthy donors, and from the glioblastoma cell line LN319 was extracted and mixed in different proportions from 0 to 100%. The LN319 cell line contains a point mutation at codon 15 exon 1 of the PTEN gene. The PCR-SSCP experiments demonstrated mutations in samples containing as little as 10% tumor DNA. In contrast, direct DNA sequencing experiments were less sensitive, requiring 30-70% of tumor DNA in the sample, depending on the DNA strand sequenced. In conclusion, PCR-SSCP, in our hands, is more sensitive than automated sequencing for detecting PTEN point mutations. We recommend to always sequence both strands, and take into account that samples containing less than 30% tumor cells should not only be sequenced, but also studied by PCR-SSCP in order to discriminate false negative results.


Subject(s)
Brain Neoplasms/genetics , DNA, Neoplasm/analysis , Glioblastoma/genetics , Mutation , Phosphoric Monoester Hydrolases/genetics , Tumor Suppressor Proteins , DNA Mutational Analysis , Humans , PTEN Phosphohydrolase , Polymerase Chain Reaction/methods , Polymorphism, Single-Stranded Conformational , Sequence Analysis, DNA , Silver Staining/methods , Spain
11.
Oncogene ; 19(22): 2687-94, 2000 May 18.
Article in English | MEDLINE | ID: mdl-10851068

ABSTRACT

LNCaP prostatic cancer cells are characterized by having a PTEN mutation, low levels of type 1 insulin-like growth factor receptor (IGF-IR) and no IRS-1, one of the major substrates of the IGF-IR. The absence of IRS-1, an activator of PI3-kinase, is compensated in these cells by the mutation in PTEN, an inhibitor of PI3-kinase. However, IGF-IR signaling in the absence of IRS-1 can cause cell differentiation and growth arrest. We hypothesized that these three characteristics may not be unrelated, specifically that, together, they may favor the metastatic spread of prostatic cancer cells without decreasing their growth potential. In support of this hypothesis, we report here that: (1) IRS-1 expression increases cell adhesion and decreases cell motility; (2) over-expression of the IGF-IR, in the absence of IRS-1, causes growth arrest and (3) a combination of IGF-IR and IRS-1 restores the transformed phenotype of LNCaP cells. These findings suggest a mechanism by which prostatic cancer cells can achieve metastatic potential without interfering with their growth potential. Oncogene (2000).


Subject(s)
Gene Expression Regulation, Neoplastic , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction/genetics , Tumor Suppressor Proteins , Animals , Cell Adhesion , Cell Movement/genetics , Genes, Tumor Suppressor , Germ-Line Mutation , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , PTEN Phosphohydrolase , Prostatic Neoplasms/pathology , Tumor Cells, Cultured
12.
Neuro Oncol ; 2(1): 6-15, 2000 01.
Article in English | MEDLINE | ID: mdl-11302255

ABSTRACT

Differential display polymerase chain reaction analysis was used to compare five differentiation states of the O-2A progenitor-like cell line CG4: progenitor cells and cells at 12 h or 4 days after the induction of differentiation into oligodendrocytes or astrocytes. This led to the identification of 52 sequence tags that were expressed differentially with cellular phenotype. One sequence was upregulated during differentiation of CG4 cells and represented a novel gene that we named SETA (SH3 domain-containing gene expressed in tumorigenic astrocytes). This gene encodes an SH3 domain-containing adapter protein with sequence similarity to the CD2AP (CD2 adapter protein) and CMS (Cas ligand with multiple Src homology) genes. SETA mRNA was expressed at high levels in the developing rat brain but was barely detectable in the normal adult rat or human brain. However, SETA mRNA was found in approximately one half of the human gliomas tested, including astrocytomas grades II, III, and IV, as well as oligodendrogliomas, mixed oligoastrocytomas, and human glioma-derived cell lines. A rat glioma generated by treatment with the alkylating carcinogen ethylnitrosourea on postnatal day 1 and a derived cell line also expressed SETA mRNA. Furthermore, in an in vitro model of astrocytoma progression based on p53-/- astrocytes, expression of SETA was restricted to cells that are tumorigenic.


Subject(s)
Astrocytes/physiology , Central Nervous System Neoplasms/etiology , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Astrocytes/cytology , Astrocytes/metabolism , Brain/embryology , Brain/growth & development , Brain/metabolism , Brain Neoplasms/metabolism , Cell Line , DNA, Complementary/genetics , Data Display , Embryonic and Fetal Development , Glioma/metabolism , Humans , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Neuroglia/cytology , Neuroglia/metabolism , Oligodendroglia/cytology , Polymerase Chain Reaction , Rats , Stem Cells/cytology
13.
Oncogene ; 18(27): 3936-43, 1999 Jul 08.
Article in English | MEDLINE | ID: mdl-10435616

ABSTRACT

The tumor suppressor gene PTEN (MMAC1, TEP1) encodes a dual-specificity phosphatase and is considered a progression-associated target of genetic alterations in human gliomas. Recently, it has been reported that the introduction of wild type PTEN into glioma cells containing endogenous mutant PTEN alleles (U87MG, LN-308), but not in those which retain wild-type PTEN (LN-18, LN-229), causes growth suppression and inhibits cellular migration, spreading and focal adhesion. Here, we show that PTEN gene transfer has no effect on the chemosensitivity of the four cell lines. Further, a correlational analysis of the endogenous PTEN status of 12 human glioma cell lines with their sensitivity to seven different cancer chemotherapy drugs reveals no link between PTEN and chemosensitivity. In contrast, ectopic expression of wild type PTEN, but not the PTEN(G129R) mutant, in PTEN-mutant gliomas markedly sensitizes these cells to irradiation and to CD95-ligand (CD95L)-induced apoptosis. PTEN-mediated facilitation of CD95L-induced apoptosis is associated with enhanced CD95L-evoked caspase 3 activity. Protein kinase B (PKB/Akt), previously shown to inhibit CD95L-induced apoptosis in nonglial COS7 cells, is inactivated by dephosphorylation. Interestingly, both PTEN-mutant U87MG and PTEN-wild-type LN-229 cells contain phosphorylated PKB constitutively. Wild-type PTEN gene transfer promotes dephosphorylation of PKB specifically in U87MG cells but not in LN-229 cells. Sensitization of U87MG cells to CD95L-apoptosis by wild-type PTEN is blocked by insulin-like growth factor-1 (IGF-1). The protection by IGF-1 is inhibited by the phosphoinositide 3-OH (PI 3) kinase inhibitor, wortmannin. Although PKB is a down-stream target of PI 3 kinase, the protection by IGF-1 was not associated with the reconstitution of PKB phosphorylation. Thus, PTEN may sensitize human malignant glioma cells to CD95L-induced apoptosis in a PI 3 kinase-dependent manner that may not require PKB phosphorylation.


Subject(s)
Apoptosis/genetics , Gene Transfer Techniques , Glioma/genetics , Membrane Glycoproteins/physiology , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/radiation effects , Tumor Suppressor Proteins , fas Receptor/physiology , Antineoplastic Agents/pharmacology , Apoptosis/radiation effects , Fas Ligand Protein , Gamma Rays , Glioma/drug therapy , Glioma/pathology , Glioma/radiotherapy , Growth Inhibitors/genetics , Growth Inhibitors/physiology , Humans , Mutation , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/biosynthesis , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology , Tumor Cells, Cultured/radiation effects
14.
Cancer Res ; 58(22): 5002-8, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9823298

ABSTRACT

The PTEN gene (also called MMAC1 and TEP1) at chromosome 10q23 is mutated in a variety of predominantly late-stage tumors and has been shown to suppress glioma cell growth in vitro and in vivo. Here we sought to determine the mechanism by which PTEN mediates growth inhibition. Using the mutant PTEN glioma cell line, U87MG, as a transfection recipient for a series of PTEN alleles, we provide direct evidence that this capacity requires phosphatase activity. Mutations mapping upstream, within, and downstream of the catalytic domain ablated activity toward a 3' phosphorylated phosphoinositide substrate of PTEN, whereas alleles with mutations flanking the catalytic domain retained activity toward the acidic protein polymer substrate, Glu4Tyr1. Thus, catalytic activity toward phosphoinositide substrates was required for growth suppression, whereas activity toward the protein substrate was dispensable for growth suppression. Finally, we used apoptotic and cell proliferation analyses to show that PTEN-mediated growth inhibition under reduced serum conditions was due to a G1 cell cycle block rather than to an induction of apoptosis.


Subject(s)
G1 Phase/genetics , Genes, Tumor Suppressor/physiology , Glioma/pathology , Phosphoric Monoester Hydrolases/genetics , Tumor Suppressor Proteins , Alleles , Apoptosis , Cell Division/genetics , Flow Cytometry , Genetic Vectors/genetics , Glioma/genetics , Humans , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/physiology , Transfection , Tumor Cells, Cultured
15.
Proc Natl Acad Sci U S A ; 95(16): 9418-23, 1998 Aug 04.
Article in English | MEDLINE | ID: mdl-9689095

ABSTRACT

Gross genetic lesions of chromosome 10 occur in 30-50% of sporadic human melanomas. To test the functional significance of this observation, we have developed an in vitro loss of heterozygosity approach in which a wild-type chromosome 10 was transferred into melanoma cells, where there was selection for its breakage and regional deletion to relieve its growth suppressive effects. The overlap of these events was at band 10q23, the site of the recently isolated PTEN/MMAC1 tumor suppressor gene, suggesting it as a potential target. Although the gene was expressed in the parental cells, both of its chromosomal alleles contained truncating mutations. In vitro loss of heterozygosity resulted in loss of the chromosomally introduced wild-type PTEN/MMAC1, and ectopic expression of the gene caused cell growth suppression. Thus, this approach identified PTEN/MMAC1 as a target in malignant melanoma and may provide an alternative means to localizing tumor suppressor genes.


Subject(s)
Loss of Heterozygosity , Melanoma/genetics , Phosphoric Monoester Hydrolases , Protein Tyrosine Phosphatases/genetics , Tumor Suppressor Proteins , Base Sequence , DNA Primers , Humans , Melanoma/pathology , PTEN Phosphohydrolase , Tumor Cells, Cultured
16.
Cancer Res ; 58(13): 2724-6, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9661881

ABSTRACT

Bannayan-Riley-Ruvalcaba syndrome (BRRS) is a rare hamartomatous polyposis condition with features of macrocephaly, intestinal juvenile polyposis, developmental delay, lipomas, and pigmentation spots of the male genitalia. An autosomal dominant pattern of inheritance exists in some families, but others appear as sporadic cases. Germ-line mutations in PTEN, a tyrosine phosphatase and putative tumor suppressor gene, have been demonstrated in two families with BRRS, and chromatin loss at the PTEN gene locus on chromosome 10q23 has been demonstrated in two BRRS patients. Germ-line mutations in PTEN have also been described in Cowden disease and in a small number of patients with juvenile polyposis syndrome. In an attempt to assess the nature of PTEN mutations in BRRS, we analyzed three sporadic BRRS patients for chromosome 10q23 deletion or PTEN germ-line mutations. All 3 patients demonstrated no loss of parental alleles at 15 chromosome 10q23 markers that encompassed the region of PTEN. In addition, analysis of mRNA and genomic DNA revealed no nonsense, missense, or insertion/deletion mutations of PTEN. Thus, other mechanisms besides mutation of PTEN must have occurred to cause BRRS in these patients. We speculate that BRRS and juvenile polyposis syndrome may have a heterogeneous etiology to cause their syndromes.


Subject(s)
Germ-Line Mutation/genetics , Hamartoma Syndrome, Multiple/genetics , Intestinal Polyps/genetics , Phosphoric Monoester Hydrolases , Pigmentation Disorders/genetics , Protein Tyrosine Phosphatases/genetics , Tumor Suppressor Proteins , DNA/genetics , Developmental Disabilities/genetics , Humans , Male , PTEN Phosphohydrolase , RNA, Messenger/genetics , Syndrome
17.
Proc Natl Acad Sci U S A ; 94(23): 12479-84, 1997 Nov 11.
Article in English | MEDLINE | ID: mdl-9356475

ABSTRACT

Deletions of all or part of chromosome 10 are the most common genetic alterations in high-grade gliomas. The PTEN gene (also called MMAC1 and TEP1) maps to chromosome region 10q23 and has been implicated as a target of alteration in gliomas and also in other cancers such as those of the breast, prostate, and kidney. Here we sought to provide a functional test of its candidacy as a growth suppressor in glioma cells. We used a combination of Northern blot analysis, protein truncation assays, and sequence analysis to determine the types and frequency of PTEN mutations in glioma cell lines so that we could define appropriate recipients to assess the growth suppressive function of PTEN by gene transfer. Introduction of wild-type PTEN into glioma cells containing endogenous mutant alleles caused growth suppression, but was without effect in cells containing endogenous wild-type PTEN. The ectopic expression of PTEN alleles, which carried mutations found in primary tumors and have been shown or are expected to inactivate its phosphatase activity, caused little growth suppression. These data strongly suggest that PTEN is a protein phosphatase that exhibits functional and specific growth-suppressing activity.


Subject(s)
Brain Neoplasms/pathology , Chromosomes, Human, Pair 10 , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Phosphoric Monoester Hydrolases , Protein Tyrosine Phosphatases/genetics , Tumor Suppressor Proteins , Brain Neoplasms/genetics , Cell Division/genetics , Humans , Mutation , PTEN Phosphohydrolase , Tumor Cells, Cultured
18.
Pediatr Neurosurg ; 24(1): 41-9, 1996.
Article in English | MEDLINE | ID: mdl-8817614

ABSTRACT

Cancer has been proposed to develop by a process of stepwise accumulation of growth-advantageous genetic alterations which result in the evolution of clones which are outgrowths of such rare cells [1]. This model has recently been extensively tested in human gliomas, the most common primary tumor of the adult central nervous system. Temporal disease progression involves an interplay between growth-suppressing and growth-promoting genes. Specifically for gliomas, genetic studies have indicated loss of germline heterozygosity for chromosome 17p; mutation of the p53 gene; overexpression of the platelet-derived growth factor-alpha receptor; allelic losses of chromosomes 22q, 13q, and 19q; deletion of the interferon-alpha and beta and CDKN2 loci on chromosome 9p; amplification and rearrangement of the epidermal growth factor receptor gene, and monosomy of chromosome 10. The following discussion details these genetic alterations and their consequences for the biology of glioma progression with the ultimate aim of providing new avenues for clinical intervention.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Glioblastoma/genetics , Adult , Astrocytoma/pathology , Brain/pathology , Brain Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Child , Chromosome Aberrations/genetics , Glioblastoma/pathology , Humans
19.
Cancer Res ; 55(9): 1941-5, 1995 May 01.
Article in English | MEDLINE | ID: mdl-7728764

ABSTRACT

P16INK4 is a cell cycle regulator that specifically binds to and inactivates cyclin-dependent kinase 4 (CDK4). Its encoding gene (p16/CDKN2) maps to chromosome 9p21, a region that undergoes frequent loss of heterozygosity in a variety of human tumors. We have analyzed the p16/CDKN2 gene and its expression in a series of primary glioma samples. Although homozygous deletion or mutation of the p16/CDKN2 gene was uncommon in this series and P16INK4 protein was detectable in all grade II tumors, it was present in only 50% of grade III and grade IV samples. Conversely, in some grade IV tumors that level of P16INK4 protein was elevated; in these cases, its target, CDK4, was amplified and overexpressed. These results suggest: (a) the involvement of P16INK4 in glioma progression; (b) that mechanisms other than mutation or deletion can down-regulate expression of the p16/CDKN2 gene; and (c) that the balance between CDK4 and its cognate inhibitor, P16INK4, may confer a cell growth advantage and facilitate tumor progression.


Subject(s)
Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Carrier Proteins/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Base Sequence , Blotting, Western , Carrier Proteins/analysis , Cyclin-Dependent Kinase Inhibitor p16 , Disease Progression , Gene Deletion , Gene Expression , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/genetics , Humans , Molecular Sequence Data , Mutation , Protein Kinase Inhibitors , RNA, Messenger/analysis , RNA, Messenger/genetics , Transcription, Genetic
20.
Cancer Res ; 55(6): 1351-4, 1995 Mar 15.
Article in English | MEDLINE | ID: mdl-7882335

ABSTRACT

The p16/CDKN2 gene has many features of a growth suppressor gene: it maps to 9p21, a frequent region of loss of heterozygozity in a variety of tumor types; it encodes an inhibitor of cyclin-dependent kinase 4; and its homozygous deletion is common in tumor-derived cell lines. However, the lower frequency of alteration of the gene in primary tumor tissue as compared to the cognate tumor cell lines has brought this interpretation into question. We have assessed the growth suppressive function of p16/CDKN2 by gene transfer. The introduction of full-length p16/CDKN2 cDNA caused marked growth suppression in p16/CDKN2-null human glioma cells, but was without significant effect in those cells with endogenous wild-type p16/CDKN2 alleles. These results provide functional evidence in support of the hypothesis that the p16/CDKN2 gene is a functional growth suppressor gene, at least in gliomas.


Subject(s)
Carrier Proteins/genetics , Genes, Tumor Suppressor , Glioma/genetics , Protein Kinase Inhibitors , Base Sequence , Cell Division , Cyclin-Dependent Kinase Inhibitor p16 , Gene Transfer Techniques , Glioma/pathology , Humans , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...