Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
3.
Proteomics ; 11(20): 4021-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21898824

ABSTRACT

We have recently demonstrated that Notch pathway blockade by γ-secretase inhibitor (GSI) depletes cancer stem cells (CSCs) in Glioblastoma Multiforme (GBM) through reduced proliferation and induced apoptosis. However, the detailed mechanism by which the manipulation of Notch signal induces alterations on post-translational modifications such as glycosylation has not been investigated. Herein, we present a differential profiling work to detect the change of glycosylation pattern upon drug treatment in GBM CSCs. Rapid screening of differential cell surface glycan structures has been performed by lectin microarray on live cells followed by the detection of N-linked glycoproteins from cell lysates using multi-lectin chromatography and label-free quantitative mass spectrometry analysis. A total of 51 and 52 glycoproteins were identified in the CSC- and GSI-treated groups, respectively, filtered by a combination of decoy database searching and Trans-Proteomic Pipeline (TPP) processing. Although no significant changes were detected from the lectin microarray experiment, 7 differentially expressed glycoproteins with high confidence were captured after the multi-lectin column including key enzymes involved in glycan processing. Functional annotations of the altered glycoproteins suggest a phenotype transformation of CSCs toward a less tumorigenic form upon GSI treatment.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Glioblastoma , Glycoproteins/chemistry , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Blotting, Western , Humans , Lectins/chemistry , Neoplastic Stem Cells/cytology , Protein Array Analysis
4.
Cancer Res ; 71(18): 6061-72, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21788346

ABSTRACT

One important function of endothelial cells in glioblastoma multiforme (GBM) is to create a niche that helps promote self-renewal of cancer stem-like cells (CSLC). However, the underlying molecular mechanism for this endothelial function is not known. Since activation of NOTCH signaling has been found to be required for propagation of GBM CSLCs, we hypothesized that the GBM endothelium may provide the source of NOTCH ligands. Here, we report a corroboration of this concept with a demonstration that NOTCH ligands are expressed in endothelial cells adjacent to NESTIN and NOTCH receptor-positive cancer cells in primary GBMs. Coculturing human brain microvascular endothelial cells (hBMEC) or NOTCH ligand with GBM neurospheres promoted GBM cell growth and increased CSLC self-renewal. Notably, RNAi-mediated knockdown of NOTCH ligands in hBMECs abrogated their ability to induce CSLC self-renewal and GBM tumor growth, both in vitro and in vivo. Thus, our findings establish that NOTCH activation in GBM CSLCs is driven by juxtacrine signaling between tumor cells and their surrounding endothelial cells in the tumor microenvironment, suggesting that targeting both CSLCs and their niche may provide a novel strategy to deplete CSLCs and improve GBM treatment.


Subject(s)
Brain Neoplasms/pathology , Endothelial Cells/pathology , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Receptors, Notch/metabolism , Stem Cell Niche , AC133 Antigen , Adaptor Proteins, Signal Transducing , Animals , Antigens, CD/metabolism , Brain Neoplasms/metabolism , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cell Growth Processes/physiology , Endothelial Cells/metabolism , Gene Knockdown Techniques , Glioblastoma/metabolism , Glycoproteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Intermediate Filament Proteins/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Peptides/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Receptors, Notch/biosynthesis , Receptors, Notch/deficiency , Receptors, Notch/genetics , Serrate-Jagged Proteins , Xenograft Model Antitumor Assays
5.
J Neurooncol ; 103(2): 247-53, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20853134

ABSTRACT

The stem cell factor/kit tyrosine kinase receptor pathway is related to tumor growth and progression in several cancers including Ewing sarcoma, a peripheral PNET (pPNET). Identifying additional groups of tumors that may use the pathway is important as they might be responsive to imatinib mesylate treatment. MB and central PNET (cPNET) are embryonal tumors of the CNS that share similar undifferentiated morphology with Ewing sarcomas and display aggressive clinical behavior. cPNET outcome is significantly lower than MB outcome, even for localized tumors treated with high-risk MB therapy. The elucidation of signaling pathways involved in MB and cPNET pathogenesis, and the discovery of new therapeutic targets is necessary to improve the treatment of these neoplasms. We analyzed KIT expression in 2 MB, one pPNET, one cPNET and 2 rhabdomyosarcoma (RMS) cell lines. Also, in 13 tumor samples (12 MB and one cPNET), we found KIT overexpression in the most aggressive cell lines (metastatic MB and pPNET). Hypermethylation of KIT was clear in the RMS non-expressing cell lines. Among MB tumors, we could see variable levels of KIT expression; a subset of them (25%) might be related in its growth pattern to KIT up-regulation. No methylated KIT was detected in the tumors expressing the lowest levels of KIT. Our results point to methylation as an epigenetic regulatory mechanism for KIT inhibition only in the KIT non-expressing RMS cell lines, and neither in the rest of the cell lines nor in the tumor samples.


Subject(s)
Cerebellar Neoplasms/genetics , DNA Methylation/genetics , Medulloblastoma/genetics , Nerve Sheath Neoplasms/genetics , Proto-Oncogene Proteins c-kit/genetics , Blotting, Western , Cell Line, Tumor , Cerebellar Neoplasms/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Medulloblastoma/metabolism , Nerve Sheath Neoplasms/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
6.
J Proteome Res ; 10(1): 330-8, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21110520

ABSTRACT

Cancer stem cells are responsible for tumor formation through self-renewal and differentiation into multiple cell types and thus represent a new therapeutic target for tumors. Glycoproteins play a critical role in determining the fates of stem cells such as self-renewal, proliferation, and differentiation. Here we applied a multilectin affinity chromatography and quantitative glycoproteomics approach to analyze alterations of glycoproteins relevant to the differentiation of a glioblastoma-derived stem cell line HSR-GBM1. Three lectins including concanavalin A (Con A), wheat germ agglutinin (WGA), and peanut agglutinin (PNA) were used to capture glycoproteins, followed by LC-MS/MS analysis. A total of 73 and 79 high-confidence (FDR < 0.01) glycoproteins were identified from the undifferentiated and differentiated cells, respectively. Label-free quantitation resulted in the discovery of 18 differentially expressed glycoproteins, wherein 9 proteins are localized in the lysosome. All of these lysosomal glycoproteins were up-regulated after differentiation, where their principal function was hydrolysis of glycosyl residues. Protein-protein interaction and functional analyses revealed the active involvement of lysosomes during the process of glioblastoma stem cell differentiation. This work provides glycoprotein markers to characterize differentiation status of glioblastoma stem cells that may be useful in stem-cell therapy of glioblastoma.


Subject(s)
Cell Differentiation , Glioblastoma/chemistry , Glycoproteins/chemistry , Proteomics/methods , Blotting, Western , Cell Line, Tumor , Cell Shape , Chromatography, Affinity , Chromatography, Liquid , Cluster Analysis , Databases, Protein , Glioblastoma/metabolism , Glycoproteins/isolation & purification , Glycoproteins/metabolism , Humans , Lysosomes/metabolism , Microscopy, Phase-Contrast , Neoplastic Stem Cells , Plant Lectins/metabolism , Protein Interaction Mapping , Tandem Mass Spectrometry
7.
Stem Cells ; 28(1): 5-16, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19904829

ABSTRACT

Cancer stem cells (CSCs) are thought to be critical for the engraftment and long-term growth of many tumors, including glioblastoma (GBM). The cells are at least partially spared by traditional chemotherapies and radiation therapies, and finding new treatments that can target CSCs may be critical for improving patient survival. It has been shown that the NOTCH signaling pathway regulates normal stem cells in the brain, and that GBMs contain stem-like cells with higher NOTCH activity. We therefore used low-passage and established GBM-derived neurosphere cultures to examine the overall requirement for NOTCH activity, and also examined the effects on tumor cells expressing stem cell markers. NOTCH blockade by gamma-secretase inhibitors (GSIs) reduced neurosphere growth and clonogenicity in vitro, whereas expression of an active form of NOTCH2 increased tumor growth. The putative CSC markers CD133, NESTIN, BMI1, and OLIG2 were reduced following NOTCH blockade. When equal numbers of viable cells pretreated with either vehicle (dimethyl sulfoxide) or GSI were injected subcutaneously into nude mice, the former always formed tumors, whereas the latter did not. In vivo delivery of GSI by implantation of drug-impregnated polymer beads also effectively blocked tumor growth, and significantly prolonged survival, albeit in a relatively small cohort of animals. We found that NOTCH pathway inhibition appears to deplete stem-like cancer cells through reduced proliferation and increased apoptosis associated with decreased AKT and STAT3 phosphorylation. In summary, we demonstrate that NOTCH pathway blockade depletes stem-like cells in GBMs, suggesting that GSIs may be useful as chemotherapeutic reagents to target CSCs in malignant gliomas.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Glioblastoma/drug therapy , Glycoproteins/metabolism , Neurons/drug effects , Peptides/metabolism , Receptor, Notch2/metabolism , Signal Transduction/drug effects , AC133 Antigen , Amyloid Precursor Protein Secretases/metabolism , Animals , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Dose-Response Relationship, Drug , Female , Glioblastoma/enzymology , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Neurons/enzymology , Neurons/immunology , Neurons/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Notch2/genetics , STAT3 Transcription Factor/metabolism , Spheroids, Cellular , Time Factors , Transfection , Tumor Burden , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...