Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci Methods ; 198(1): 99-102, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21402102

ABSTRACT

Isolation of glioblastoma stem cells requires incubation of tumor cells in a neural stem cell media. Neurospheres containing these glioblastoma stem cells are formed after approximately a five-day period. These cells can then be analyzed for the presence of stem cell markers. Immunofluorescence staining for these markers can serve as a valuable tool for analyzing the intact neurosphere directly in stem cell media. Here we present the use of a novel fixative (1,4-benzoquinone) for immunoflourescence staining of neurospheres.


Subject(s)
Antigens, CD/metabolism , Glycoproteins/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Peptides/metabolism , AC133 Antigen , Animals , Benzoquinones/pharmacology , Fixatives/pharmacology , Glioblastoma/pathology , Intermediate Filament Proteins/metabolism , Mice , Neoplastic Stem Cells/drug effects , Nerve Tissue Proteins/metabolism , Nestin , Tumor Cells, Cultured
2.
J Neuropathol Exp Neurol ; 69(6): 582-92, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20467333

ABSTRACT

One of the major pathophysiological features of malignant astrocytomas is their ability to infiltrate surrounding brain tissue. The epidermal growth factor receptor (EGFR) and proteases are known to be overexpressed in glioblastomas (GBMs), but the interaction between the activation of the EGFR and urokinase plasminogen activator (uPA) in promoting astrocytic tumor invasion has not been fully elucidated. Here, we characterized the signal transduction pathway(s) by which EGF regulates uPA expression and promotes astrocytoma invasion. We show that EGFR activation and constitutively active EGFR vIII in GBM cell lines upregulate uPA expression. Small-molecule inhibitors of mitogen-activated protein kinase, tyrosine kinase, and small interfering RNA targeting c-Src blocked uPA upregulation. Similarly, mutations in the activator protein 1 binding site of the uPA promoter reduced EGF-induced increases in uPA promoter activity. Treatment of GBM cells with EGF increased in vitro cell invasion, and the invasive phenotype was attenuated by gene silencing of uPA using small interfering RNA and short hairpin RNA. In addition, uPA knockdown clones formed smaller well-circumscribed tumors than nontarget U1242 control cells in a xenograft GBM mouse model in vivo. In summary, these results suggest that c-Src, mitogen-activated protein kinase, and a composite activator protein 1 on the uPA promoter are responsible for EGF-induced uPA expression and GBM invasion.


Subject(s)
Cell Movement/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Glioblastoma/metabolism , MAP Kinase Signaling System/drug effects , Transcription Factor AP-1/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Northern , Blotting, Western , Cell Line , Cells, Cultured , ErbB Receptors/genetics , Humans , Image Processing, Computer-Assisted , MAP Kinase Signaling System/physiology , Magnetic Resonance Imaging , Mice , Promoter Regions, Genetic/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Time Factors , Transcription Factor AP-1/genetics , Transfection , Transplantation, Heterologous , Urokinase-Type Plasminogen Activator/genetics
3.
Am J Pathol ; 176(6): 3032-49, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20413683

ABSTRACT

The lack of an intracranial human glioma model that recapitulates the extensive invasive and hypervascular features of glioblastoma (GBM) is a major hurdle for testing novel therapeutic approaches against GBM and studying the mechanism of GBM invasive growth. We characterized a high matrix metalloproteinase-9 (MMP-9) expressing U1242 MG intracranial xenograft mouse model that exhibited extensive individual cells and cell clusters in a perivascular and subpial cellular infiltrative pattern, geographic necrosis and infiltrating tumor-induced vascular proliferation closely resembling the human GBM phenotype. MMP-9 silencing cells with short hairpin RNA dramatically blocked the cellular infiltrative pattern, hypervascularity, and cell proliferation in vivo, and decreased cell invasion, colony formation, and cell motility in vitro, indicating that a high level of MMP-9 plays an essential role in extensive infiltration and hypervascularity in the xenograft model. Moreover, epidermal growth factor (EGF) failed to stimulate MMP-9 expression, cell invasion, and colony formation in MMP-9-silenced clones. An EGF receptor (EGFR) kinase inhibitor, a RasN17 dominant-negative construct, MEK and PI3K inhibitors significantly blocked EGF/EGFR-stimulated MMP-9, cell invasion, and colony formation in U1242 MG cells, suggesting that MMP-9 is involved in EGFR/Ras/MEK and PI3K/AKT signaling pathway-mediated cell invasion and anchorage-independent growth in U1242 MG cells. Our data indicate that the U1242 MG xenograft model is valuable for studying GBM extensive invasion and angiogenesis as well as testing anti-invasive and anti-angiogenic therapeutic approaches.


Subject(s)
Brain Neoplasms , Disease Models, Animal , Glioblastoma , Matrix Metalloproteinase 9/metabolism , Transplantation, Heterologous , Animals , Astrocytes/cytology , Astrocytes/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cell Movement , Cell Proliferation , Cells, Cultured , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Male , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/physiology , ras Proteins/metabolism
4.
J Neurosci ; 29(14): 4605-15, 2009 Apr 08.
Article in English | MEDLINE | ID: mdl-19357285

ABSTRACT

MMPs (matrix metalloproteinases) and the related "a disintegrin and metalloproteinases" (ADAMs) promote tumorigenesis by cleaving extracellular matrix and protein substrates, including N-cadherin. Although N-cadherin is thought to regulate cell adhesion, migration, and invasion, its role has not been characterized in glioblastomas (GBMs). In this study, we investigated the expression and function of posttranslational N-cadherin cleavage in GBM cells as well as its regulation by protein kinase C (PKC). N-Cadherin cleavage occurred at a higher level in glioblastoma cells than in non-neoplastic astrocytes. Treatment with the PKC activator phorbol 12-myristate 13-acetate (PMA) increased N-cadherin cleavage, which was reduced by pharmacological inhibitors and short interfering RNA (siRNA) specific for ADAM-10 or PKC-alpha. Furthermore, treatment of GBM cells with PMA induced the translocation of ADAM-10 to the cell membrane, the site at which N-cadherin was cleaved, and this translocation was significantly reduced by the PKC-alpha inhibitor Gö6976 [12-(2-cyanoethyl)-6,7,12,13-tetrahydro-13-methyl-5-oxo-5H-indolo[2,3-a]pyrrolo[3,4-c]carbazole] or PKC-alpha short hairpin RNA. In functional studies, N-cadherin cleavage was required for GBM cell migration, as depletion of N-cadherin cleavage by N-cadherin siRNA, ADAM-10 siRNA, or a cleavage-site mutant N-cadherin, decreased GBM cell migration. Together, these results suggest that N-cadherin cleavage is regulated by a PKC-alpha-ADAM-10 cascade in GBM cells and may be involved in mediating GBM cell migration.


Subject(s)
ADAM Proteins/physiology , Amyloid Precursor Protein Secretases/physiology , Antigens, CD/metabolism , Cadherins/metabolism , Cell Movement/physiology , Glioblastoma/enzymology , Membrane Proteins/physiology , Protein Kinase C-alpha/physiology , ADAM Proteins/chemistry , ADAM10 Protein , Amyloid Precursor Protein Secretases/chemistry , Antigens, CD/chemistry , Cadherins/chemistry , Cell Line, Tumor , Cell Migration Inhibition/genetics , Cell Migration Inhibition/physiology , Cell Movement/genetics , Cells, Cultured , Glioblastoma/pathology , Humans , Hydrolysis , Membrane Proteins/chemistry , Mutation
5.
Glia ; 56(8): 917-24, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18383343

ABSTRACT

Previous study reported that the activation of Ras pathway cooperated with E6/E7-mediated inactivation of p53/pRb to transform immortalized normal human astrocytes (NHA/hTERT) into intracranial tumors strongly resembling human astrocytomas. The mechanism of how H-Ras contributes to astrocytoma formation is unclear. Using genetically modified NHA cells (E6/E7/hTERT and E6/E7/hTERT/Ras cells) as models, we investigated the mechanism of Ras-induced tumorigenesis. The overexpression of constitutively active H-RasV12 in E6/E7/hTERT cells robustly increased the levels of urokinase plasminogen activator (uPA) mRNA, protein, activity and invasive capacity of the E6/E7/hTERT/Ras cells. However, the expressions of MMP-9 and MMP-2 did not significantly change in the E6/E7/hTERT and E6/E7/hTERT/Ras cells. Furthermore, E6/E7/hTERT/Ras cells also displayed higher level of uPA activity and were more invasive than E6/E7/hTERT cells in 3D culture, and formed an intracranial tumor mass in a NOD-SCID mouse model. uPA specific inhibitor (B428) and uPA neutralizing antibody decreased uPA activity and invasion in E6/E7/hTERT/Ras cells. uPA-deficient U-1242 glioblastoma cells were less invasive in vitro and exhibited reduced tumor growth and infiltration into normal brain in xenograft mouse model. Inhibitors of Ras (FTA), Raf (Bay 54-9085) and MEK (UO126), but not of phosphatidylinositol 3-kinase (PI3K) (LY294002) and of protein kinase C (BIM) pathways, inhibited uPA activity and cell invasion. Our results suggest that H-Ras increased uPA expression and activity via the Ras/Raf/MEK signaling pathway leading to enhanced cell invasion and this may contribute to increased invasive growth properties of astrocytomas.


Subject(s)
Astrocytes/physiology , Mitogen-Activated Protein Kinases/physiology , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/physiology , Urokinase-Type Plasminogen Activator/metabolism , Analysis of Variance , Animals , Brain Neoplasms/pathology , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/physiology , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Transfection/methods , ras Proteins/metabolism
6.
Cancer Res ; 67(21): 10241-51, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17974965

ABSTRACT

Aggressive and infiltrative invasion is one of the hallmarks of glioblastoma. Low-density lipoprotein receptor-related protein (LRP) is expressed by glioblastoma, but the role of this receptor in astrocytic tumor invasion remains poorly understood. We show that activation of protein kinase C-alpha (PKC-alpha) phosphorylated and down-regulated LRP expression. Pretreatment of tumor cells with PKC inhibitors, phosphoinositide 3-kinase (PI3K) inhibitor, PKC-alpha small interfering RNA (siRNA), and short hairpin RNA abrogated phorbol 12-myristate 13-acetate-induced down-regulation of LRP and inhibited astrocytic tumor invasion in vitro. In xenograft glioblastoma mouse model and in vitro transmembrane invasion assay, LRP-deficient cells, which secreted high levels of urokinase-type plasminogen activator (uPA), invaded extensively the surrounding normal brain tissue, whereas the LRP-overexpressing and uPA-deficient cells did not invade into the surrounding normal brain. siRNA, targeted against uPA in LRP-deficient clones, attenuated their invasive potential. Taken together, our results strongly suggest the involvement of PKC-alpha/PI3K signaling pathways in the regulation of LRP-mediated astrocytoma invasion. Thus, a strategy of combining small molecule inhibitors of PKC-alpha and PI3K could provide a new treatment paradigm for glioblastomas.


Subject(s)
Astrocytoma/pathology , Low Density Lipoprotein Receptor-Related Protein-1/physiology , Protein Kinase C-alpha/physiology , Urokinase-Type Plasminogen Activator/physiology , Animals , Astrocytoma/therapy , Cell Line, Tumor , Cell Movement , Humans , Immunoprecipitation , Male , Mice , Mice, SCID , Neoplasm Invasiveness , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C-alpha/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology
7.
Am J Pathol ; 170(1): 356-65, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17200207

ABSTRACT

The complete resection of pituitary adenomas (PAs) is unlikely when there is an extensive local dural invasion and given that the molecular mechanisms remain primarily unknown. DNA microarray analysis was performed to identify differentially expressed genes between nonfunctioning invasive and noninvasive PAs. Gene clustering revealed a robust eightfold increase in matrix metalloproteinase (MMP)-9 expression in surgically resected human invasive PAs and in the (nonfunctioning) HP75 human pituitary tumor-derived cell line treated with phorbol-12-myristate-13-acetate; these results were confirmed by real-time polymerase chain reaction, gelatin zymography, reverse transcriptase-polymerase chain reaction, Western blot, immunohistochemistry, and Northern blot analyses. The activation of protein kinase C (PKC) increased both MMP-9 activity and expression, which were blocked by some PKC inhibitors (Gö6976, bisindolylmaleimide, and Rottlerin), PKC-alpha, and PKC-delta small interfering (si)RNAs but not by hispidin (PKC-beta inhibitor). In a transmembrane invasion assay, phorbol-12-myristate-13-acetate (100 nmol/L) increased the number of invaded HP75 cells, a process that was attenuated by PKC inhibitors, MMP-9 antibody, PKC-alpha siRNA, or PKC-delta siRNA. These results demonstrate that MMP-9 and PKC-alpha or PKC-delta may provide putative therapeutic targets for the control of PA dural invasion.


Subject(s)
Adenoma , Matrix Metalloproteinase 9/genetics , Neoplasm Invasiveness/genetics , Pituitary Neoplasms , Adenoma/enzymology , Adenoma/pathology , Cell Line, Tumor , Enzyme Activation , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Matrix Metalloproteinase 9/biosynthesis , Pituitary Neoplasms/enzymology , Pituitary Neoplasms/pathology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Pyrones/pharmacology , RNA, Small Interfering , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology
8.
J Neurosci Methods ; 150(2): 265-71, 2006 Jan 30.
Article in English | MEDLINE | ID: mdl-16118020

ABSTRACT

The mitochondrial DNA (mtDNA) depletion status of rho(0) cell lines is typically assessed by hybridization or polymerase chain reaction (PCR) experiments, in which the failure to hybridize mtDNA or amplify mtDNA using mtDNA-directed primers suggests thorough mitochondrial genome removal. Here, we report the use of an mtDNA pseudogene ratioing technique for the additional confirmation of rho0 status. Total genomic DNA from a U251 human glioma cell line treated with ethidium bromide was amplified using primers designed to anneal either mtDNA or a previously described nuclear DNA-embedded mtDNA pseudogene (mtDNApsi). The resultant PCR product was used to generate plasmid clones. Sixty-two plasmid clones were genotyped, and all arose from mtDNApsi template. These data allowed us to determine with 95% confidence that the resultant mtDNA-depleted cell line contains less than one copy of mtDNA per 10 cells. Unlike previous hybridization or PCR-based analyses of mtDNA depletion, this mtDNApsi ratioing technique does not rely on interpretation of a negative result, and may prove useful as an adjunct for the determination of rho0 status or mtDNA copy number.


Subject(s)
DNA, Mitochondrial/analysis , Nucleic Acid Amplification Techniques , Pseudogenes , Cell Line, Tumor , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/genetics , Enzyme Inhibitors/pharmacology , Ethidium/pharmacology , Gene Dosage , Humans , Polymerase Chain Reaction , Sensitivity and Specificity
9.
Neuro Oncol ; 4(1): 9-21, 2002 01.
Article in English | MEDLINE | ID: mdl-11772428

ABSTRACT

Both increased cell proliferation and apoptosis play important roles in the malignant growth of glioblastomas. We have demonstrated recently that the differential expression of protein kinase C (PKC)-eta increases the proliferative capacity of glioblastoma cells in culture; however, specific functions for this novel PKC isozyme in the regulation of apoptosis in these tumors has not been defined. In the present study of several glioblastoma cell lines, we investigated the role of PKC-eta in preventing UV- and gamma-irradiation-induced apoptosis and in caspase-dependent signaling pathways that mediate cell death. Exposure to UV or gamma irradiation killed 80% to 100% of PKC-eta-deficient nonneoplastic human astrocytes and U-1242 MG cells, but had little effect on the PKC-eta-expressing U-251 MG and U-373 MG cells. PKC-eta appears to mediate resistance to irradiation specifically such that when PKC-eta was stably expressed in U-1242 MG cells, more than 80% of these cells developed resistance to irradiation-induced apoptosis. Reducing PKC-eta expression by transient and stable expression of antisense PKC-eta in wild-type U-251 MG cells results in increased sensitivity to UV irradiation in a fashion similar to U-1242 MG cells and nonneoplastic astrocytes. Irradiation of PKC-eta-deficient glioblastoma cells resulted in the activation of caspase-9 and caspase-3, cleavage of poly (ADP-ribose) polymerase (PARP), and a substantial increase in subdiploid DNA content that did not occur in PKC-eta-expressing tumor cells. A specific inhibitor (Ac-DEVD-CHO) of caspase-3 blocked apoptosis in PKC-eta-deficient U-1242 MG cells. The data demonstrate that resistance to UV and gamma irradiation in glioblastoma cell lines is modified significantly by PKC-eta expression and that PKC-eta appears to block the apoptotic cascade at caspase-9 activation.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Enzyme Activation/radiation effects , Gamma Rays , Glioblastoma/physiopathology , Isoenzymes/physiology , Protein Kinase C/physiology , Ultraviolet Rays , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/radiation effects , Caspase 9 , Coloring Agents , DNA/metabolism , Humans , Isoenzymes/deficiency , Isoenzymes/genetics , Isoenzymes/pharmacology , Propidium , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase C/pharmacology , Staining and Labeling , Transfection , Tumor Cells, Cultured/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...