Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Neoplasia ; 17(6): 481-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26152356

ABSTRACT

Second mitochondria-derived activator of caspase (Smac) mimetics are considered as promising anticancer therapeutics that are currently under investigation in early clinical trials. They induce apoptosis by antagonizing inhibitor of apoptosis proteins, which are frequently overexpressed in cancer. We previously reported that Smac mimetics, such as BV6, additionally exert non-apoptotic functions in glioblastoma (GBM) cells by stimulating migration and invasion in a nuclear factor kappa B (NF-κB)-dependent manner. Because NF-κB target genes mediating these effects are largely unknown, we performed whole-genome expression analyses. Here, we identify chemokine (C-C motif) ligand 2 (CCL2) as the top-listed NF-κB-regulated gene being upregulated upon BV6 treatment in GBM cells. BV6-induced upregulation and secretion of CCL2 are required for migration and invasion of GBM cells because knockdown of CCL2 in GBM cells abolishes these effects. Co-culture experiments of GBM cells with non-malignant astroglial cells reveal that BV6-stimulated secretion of CCL2 by GBM cells into the supernatant triggers migration of astroglial cells toward GBM cells because CCL2 knockdown in BV6-treated GBM cells impedes BV6-stimulated migration of astroglial cells. In conclusion, we identify CCL2 as a BV6-induced NF-κB target gene that triggers migration and invasion of GBM cells and exerts paracrine effects on the GBM's microenvironment by stimulating migration of astroglial cells. These findings provide novel insights into the biological functions of Smac mimetics with important implications for the development of Smac mimetics as cancer therapeutics.


Subject(s)
Cell Movement/drug effects , Chemokine CCL2/metabolism , Glioblastoma/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondrial Proteins/metabolism , Oligopeptides/pharmacology , Tumor Microenvironment/drug effects , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Biomimetics , Blotting, Western , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Chemokine CCL2/genetics , Coculture Techniques , Gene Expression Profiling , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Paracrine Communication , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
2.
Acta Neuropathol ; 122(2): 241-51, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21590492

ABSTRACT

The suppressor of cytokine signaling 3 (SOCS3) gene is one of eight structurally related genes of the SOCS family and has been suggested to function as a tumor suppressor by inhibition of the JAK/STAT signaling pathway. We investigated 60 human gliomas of different histological types for SOCS3 alterations and found frequent SOCS3 promoter hypermethylation and transcriptional downregulation. However, SOCS3 promoter hypermethylation was virtually absent in primary glioblastomas, which are characterized by frequent epidermal growth factor receptor (EGFR) amplification and overexpression. Assessment of the relationship between SOCS3 and EGFR aberrations revealed that SOCS3 promoter hypermethylation was inversely related to both the EGFR gene dosage as well as the EGFR protein expression, thus suggesting SOCS3 inactivation as a mechanism substituting for EGFR activation in a subset of gliomas. In support of this hypothesis, stable shRNA-mediated SOCS3 knock-down in U251 glioblastoma cells resulted in an activation of EGFR-related signaling pathways, i.e. an increase in the activation levels of STAT3, FAK and to a lesser extent MAPK, while the AKT phosphorylation levels remained unaffected. Functionally, SOCS3-depletion caused strongly increased tumor cell invasion with no obvious effect on tumor cell proliferation. In summary, our findings suggest that SOCS3 inactivation by promoter hypermethylation is mutually exclusive to EGFR activation in gliomas and preferentially promotes glioma cell invasion through STAT3 and FAK activation.


Subject(s)
Brain Neoplasms/genetics , Focal Adhesion Kinase 1/metabolism , Glioma/genetics , Neoplasm Invasiveness/genetics , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , DNA Methylation/genetics , DNA Mutational Analysis , Enzyme Activation/physiology , Gene Amplification , Genes, erbB-1 , Glioma/metabolism , Glioma/pathology , Humans , Immunohistochemistry , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...