Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer ; 11: 65, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22966858

ABSTRACT

BACKGROUND: The invasion of glioblastoma cells into regions of the normal brain is a critical factor that limits current therapies for malignant astrocytomas. Previous work has identified roles for the Rho family guanine nucleotide exchange factors Trio and Vav3 in glioblastoma invasion. Both Trio and Vav3 act on the small GTPase RhoG. We therefore examined the role of RhoG in the invasive behavior of glioblastoma cells. RESULTS: We found that siRNA-mediated depletion of RhoG strongly inhibits invasion of glioblastoma cells through brain slices ex vivo. In addition, depletion of RhoG has a marginal effect on glioblastoma cell proliferation, but significantly inhibits glioblastoma cell survival in colony formation assays. We also observed that RhoG is activated by both HGF and EGF, two factors that are thought to be clinically relevant drivers of glioblastoma invasive behavior, and that RhoG is overexpressed in human glioblastoma tumors versus non-neoplastic brain. In search of a mechanism for the contribution of RhoG to the malignant behavior of glioblastoma cells, we found that depletion of RhoG strongly inhibits activation of the Rac1 GTPase by both HGF and EGF. In line with this observation, we also show that RhoG contributes to the formation of lamellipodia and invadopodia, two functions that have been shown to be Rac1-dependent. CONCLUSIONS: Our functional analysis of RhoG in the context of glioblastoma revealed a critical role for RhoG in tumor cell invasion and survival. These results suggest that targeting RhoG-mediated signaling presents a novel avenue for glioblastoma therapy.


Subject(s)
Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Glioblastoma/enzymology , Glioblastoma/pathology , rho GTP-Binding Proteins/metabolism , Animals , Brain Neoplasms/chemistry , Brain Neoplasms/metabolism , Cell Growth Processes/physiology , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Glioblastoma/chemistry , Glioblastoma/metabolism , Humans , Mice , Neoplasm Invasiveness , Neuropeptides/metabolism , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Putamen/chemistry , Putamen/metabolism , RNA, Small Interfering/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein , rho GTP-Binding Proteins/analysis , rho GTP-Binding Proteins/genetics
2.
Mol Cancer Res ; 10(7): 958-68, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22571869

ABSTRACT

Malignant glioblastomas are characterized by their ability to infiltrate into normal brain. We previously reported that binding of the multifunctional cytokine TNF-like weak inducer of apoptosis (TWEAK) to its receptor fibroblast growth factor-inducible 14 (Fn14) induces glioblastoma cell invasion via Rac1 activation. Here, we show that Cdc42 plays an essential role in Fn14-mediated activation of Rac1. TWEAK-treated glioma cells display an increased activation of Cdc42, and depletion of Cdc42 using siRNA abolishes TWEAK-induced Rac1 activation and abrogates glioma cell migration and invasion. In contrast, Rac1 depletion does not affect Cdc42 activation by Fn14, showing that Cdc42 mediates TWEAK-stimulated Rac1 activation. Furthermore, we identified two guanine nucleotide exchange factors (GEF), Ect2 and Trio, involved in TWEAK-induced activation of Cdc42 and Rac1, respectively. Depletion of Ect2 abrogates both TWEAK-induced Cdc42 and Rac1 activation, as well as subsequent TWEAK-Fn14-directed glioma cell migration and invasion. In contrast, Trio depletion inhibits TWEAK-induced Rac1 activation but not TWEAK-induced Cdc42 activation. Finally, inappropriate expression of Fn14 or Ect2 in mouse astrocytes in vivo using an RCAS vector system for glial-specific gene transfer in G-tva transgenic mice induces astrocyte migration within the brain, corroborating the in vitro importance of the TWEAK-Fn14 signaling cascade in glioblastoma invasion. Our results suggest that the TWEAK-Fn14 signaling axis stimulates glioma cell migration and invasion through two GEF-GTPase signaling units, Ect2-Cdc42 and Trio-Rac1. Components of the Fn14-Rho GEF-Rho GTPase signaling pathway present innovative drug targets for glioma therapy.


Subject(s)
Glioblastoma , Proto-Oncogene Proteins , Receptors, Tumor Necrosis Factor , cdc42 GTP-Binding Protein , Animals , Astrocytoma/genetics , Astrocytoma/metabolism , Cell Movement/genetics , Cytokine TWEAK , Glioblastoma/genetics , Glioblastoma/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Humans , Mice , Neoplasm Invasiveness/genetics , Neuropeptides/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , TWEAK Receptor , Tumor Necrosis Factors/pharmacology , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
3.
Brain Pathol ; 20(1): 17-27, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19371355

ABSTRACT

Malignant gliomas, especially glioblastomas, are associated with a dismal prognosis. Despite advances in diagnosis and treatment, glioblastoma patients still have a median survival expectancy of only 14 months. This poor prognosis can be at least partly explained by the fact that glioma cells diffusely infiltrate the brain parenchyma and exhibit decreased levels of apoptosis, and thus resistance to cytotoxic drugs. Galectins are a family of mammalian beta-galactoside-binding proteins characterized by a shared characteristic amino acid sequence. They are expressed differentially in normal vs. neoplastic tissues and are known to play important roles in several biological processes such as cell proliferation, death and migration. This review focuses on the role played by galectins, especially galectin-1 and galectin-3, in glioma biology. The involvement of these galectins in different steps of glioma malignant progression such as migration, angiogenesis or chemoresistance makes them potentially good targets for the development of new drugs to combat these malignant tumors.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Galectins/genetics , Galectins/physiology , Glioma/metabolism , Glioma/pathology , Animals , Cell Hypoxia , Cell Movement , Humans , Integrins/genetics , Integrins/physiology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology
4.
Brain Pathol ; 20(1): 39-49, 2010 Jan.
Article in English | MEDLINE | ID: mdl-18947333

ABSTRACT

Cell motility and resistance to apoptosis characterize glioblastoma (GBM) growth and malignancy. In our current work we report that galectin-1, a homodimeric adhesion molecule and carbohydrate-binding protein with affinity for beta-galactosides, is linked with cell surface expression of integrin beta1 and the process of integrin trafficking. Using immunofluorescence, depletion of galectin-1 through both stable knockdown and transient-targeted small interfering RNA (siRNA) treatment induces an intracellular accumulation of integrin-beta1 coincident with a diminution of integrin-beta1 at points of cellular adhesion at the cell membrane. Galectin-1 depletion does not alter the gene expression level of integrin-beta1. Transient galectin-1 depletion effectuates as well the perinuclear accumulation of protein kinase C epsilon (PKCepsilon) and the intermediate filament vimentin, both of which have been shown to mediate integrin recycling in motile cells. Our results argue for the involvement of galectin-1 in the PKCepsilon/vimentin-controlled trafficking of integrin-beta1. The understanding of molecular mediators such as galectin-1 and the pathways through which they drive the cell invasion so descriptive of GBM is anticipated to reveal potential therapeutic targets that promote glioma malignancy.


Subject(s)
Brain Neoplasms/metabolism , Galectin 1/genetics , Galectin 1/physiology , Glioblastoma/metabolism , Integrin beta1/biosynthesis , Integrin beta1/genetics , Protein Kinase C-epsilon/physiology , Vimentin/physiology , Antisense Elements (Genetics) , Blotting, Western , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Gene Silencing , Genomics , Humans , Integrin alpha Chains/biosynthesis , Neoplasm Proteins/analysis , Neoplasm Proteins/biosynthesis , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transfection
5.
Mol Cancer Res ; 7(11): 1871-81, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19861406

ABSTRACT

Malignant gliomas are the most common primary brain tumors. Despite intensive clinical investigation and significant technical advances in surgical and radiation treatment, the impact on clinical outcome for patients with malignant gliomas is disappointing. We have previously shown that tumor necrosis factor-like weak inducer of apoptosis (TWEAK), a member of the tumor necrosis factor superfamily, can stimulate glioma cell survival via binding to the Fn14 receptor, activation of the NF-kappaB pathway, and upregulation of BCL-X(L) gene expression. Here, we show that TWEAK treatment of glioma cells leads to phosphorylation of Akt and BAD. TWEAK stimulation results in the phosphorylation of both Akt1 and Akt2. However, small interfering RNA (siRNA)-mediated depletion of either Akt1 or Akt2 showed that BAD serine 136 phosphorylation is dependent specifically on Akt2 function. Depletion of Akt2 expression by siRNA also abrogates TWEAK-stimulated glioma cell survival, whereas no effect on glioma cell survival was observed after siRNA-mediated depletion of Akt1 expression. Surprisingly, although siRNA-mediated depletion of BAD in glioma cells abrogates cytotoxic- and chemotherapy-induced apoptosis, TWEAK still displays a strong protective effect, suggesting that BAD serine 136 phosphorylation plays a minor role in TWEAK-Akt2-induced glioma cell survival. We also report here that AKT2 gene expression levels increased with glioma grade and inversely correlate with patient survival. Additionally, immunohistochemical analysis showed that Akt2 expression positively correlates with Fn14 expression in glioblastoma multiforme specimens. We hypothesize that the TWEAK-Fn14 signaling axis functions, in part, to enhance glioblastoma cell survival by activation of the Akt2 serine/threonine protein kinase.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioma/metabolism , Glioma/pathology , Proto-Oncogene Proteins c-akt/metabolism , Tumor Necrosis Factors/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cytokine TWEAK , Gene Expression Profiling , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Glioma/drug therapy , Glioma/genetics , Humans , Immunohistochemistry , Phosphorylation , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Receptors, Tumor Necrosis Factor/metabolism , TWEAK Receptor , Transfection , Tumor Necrosis Factors/metabolism , bcl-Associated Death Protein/deficiency , bcl-Associated Death Protein/genetics
6.
Neoplasia ; 11(5): 485-96, 2009 May.
Article in English | MEDLINE | ID: mdl-19412433

ABSTRACT

We have previously reported that galectin 1 (Gal-1) plays important biological roles in astroglial as well as in oligodendroglial cancer cells. As an oligodendroglioma model, we make use of the Hs683 cell line that has been previously extensively characterized at cell biology, molecular biology, and genetic levels. Galectin 1 has been shown to be involved in Hs683 oligodendroglioma chemoresistance, neoangiogenesis, and migration. Down-regulating Gal-1 expression in Hs683 cells through targeted small interfering RNA provokes a marked decrease in the expression of the brain-expressed X-linked gene: BEX2. Accordingly, the potential role of BEX2 in Hs683 oligodendroglioma cell biology has been investigated. The data presented here reveal that decreasing BEX2 expression in Hs683 cells increases the survival of Hs683 orthotopic xenograft-bearing mice. Furthermore, this decrease in BEX2 expression impairs vasculogenic mimicry channel formation in vitro and angiogenesis in vivo, and modulates glioma cell adhesion and invasive features through the modification of several genes previously reported to play a role in cancer cell migration, including MAP2, plexin C1, SWAP70, and integrin beta(6). We thus conclude that BEX2 is implicated in oligodendroglioma biology.


Subject(s)
Cell Movement/physiology , Galectin 1/metabolism , Neovascularization, Pathologic/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglioma/metabolism , Animals , Cell Line, Tumor , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neovascularization, Pathologic/genetics , Nerve Tissue Proteins/genetics , Neurogenesis/physiology , Oligodendroglioma/genetics , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenograft Model Antitumor Assays
7.
Mol Cancer Res ; 6(5): 725-34, 2008 May.
Article in English | MEDLINE | ID: mdl-18505918

ABSTRACT

Genomic characterization is beginning to define a molecular taxonomy for breast cancer; however, the molecular basis of invasion and metastasis remains poorly understood. We report a pivotal role for the fibroblast growth factor-inducible 14 (Fn14) receptor in this process. We examined whether Fn14 and its ligand tumor necrosis factor-like weak inducer of apoptosis (TWEAK) were expressed in breast tumors and whether deregulation of Fn14 levels affected malignant behavior of breast cancer cell lines. Analysis of TWEAK and Fn14 in publicly available gene expression data indicated that high Fn14 expression levels significantly correlated with several poor prognostic indicators (P < 0.05). Fn14 expression was highest in the HER2-positive/estrogen receptor-negative (HER2(+)/ER(-)) intrinsic subtype (P = 0.0008). An association between Fn14 and HER2 expression in breast tumors was confirmed by immunohistochemistry. Fn14 levels were elevated in invasive, ER(-) breast cancer cell lines. Overexpression of Fn14 in weakly invasive MCF7 and T47D cells resulted in a marked induction of invasion and activation of nuclear factor-kappaB (NF-kappaB) signaling. Ectopic expression of Fn14tCT, a Fn14 deletion mutant that cannot activate NF-kappaB signaling, was not able to induce invasion. Moreover, ectopic expression of Fn14tCT in highly invasive MDA-MB-231 cells reduced their invasive capability. RNA interference-mediated inhibition of Fn14 expression in both MDA-MB-231 and MDA-MB-436 cells reduced invasion. Expression profiling of the Fn14-depleted cells revealed deregulation of NF-kappaB activity. Our findings support a role for Fn14-mediated NF-kappaB pathway activation in breast tumor invasion and metastasis.


Subject(s)
Breast Neoplasms/metabolism , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Neoplastic , Receptor, ErbB-2/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Adenoviridae/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , NF-kappa B/metabolism , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Prognosis , Recombinant Proteins/chemistry , TWEAK Receptor
8.
Cancer Res ; 66(19): 9535-42, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-17018610

ABSTRACT

Glial tumors progress to malignant grades by heightened proliferation and relentless dispersion throughout the central nervous system. Understanding genetic and biochemical processes that foster these behaviors is likely to reveal specific and effective targets for therapeutic intervention. Our current report shows that the fibroblast growth factor-inducible 14 (Fn14), a member of the tumor necrosis factor (TNF) receptor superfamily, is expressed at high levels in migrating glioma cells in vitro and invading glioma cells in vivo. Forced Fn14 overexpression stimulates glioma cell migration and invasion, and depletion of Rac1 by small interfering RNA inhibits this cellular response. Activation of Fn14 signaling by the ligand TNF-like weak inducer of apoptosis (TWEAK) stimulates migration and up-regulates expression of Fn14; this TWEAK effect requires Rac1 and nuclear factor-kappaB (NF-kappaB) activity. The Fn14 promoter region contains NF-kappaB binding sites, which mediate positive feedback causing sustained overexpression of Fn14 and enduring glioma cell invasion. Furthermore, Fn14 gene expression levels increase with glioma grade and inversely correlate with patient survival. These results show that the Fn14 cascade operates as a positive feedback mechanism for elevated and sustained Fn14 expression. Such a feedback loop argues for aggressive targeting of the Fn14 axis as a unique and specific driver of glioma malignant behavior.


Subject(s)
Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic/physiology , Glioma/pathology , NF-kappa B/physiology , Neoplasm Invasiveness/physiopathology , Neoplasm Proteins/physiology , Receptors, Tumor Necrosis Factor/physiology , rac1 GTP-Binding Protein/physiology , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Cell Line, Tumor/physiology , Cell Movement/genetics , Cell Movement/physiology , Feedback, Physiological , Gene Expression Regulation, Neoplastic/genetics , Glioma/metabolism , Glioma/mortality , Humans , I-kappa B Kinase/physiology , Neoplasm Invasiveness/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Prognosis , Promoter Regions, Genetic/genetics , RNA, Small Interfering/pharmacology , Rats , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/genetics , TWEAK Receptor , Transfection , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...