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
2.
Mol Carcinog ; 52(2): 118-33, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22086447

ABSTRACT

Glioblastomas are invasive tumors with poor prognosis despite current therapies. Histone deacetylase inhibitors (HDACIs) represent a class of agents that can modulate gene expression to reduce tumor growth, and we and others have noted some antiglioma activity from HDACIs, such as vorinostat, although insufficient to warrant use as monotherapy. We have recently demonstrated that proteasome inhibitors, such as bortezomib, dramatically sensitized highly resistant glioma cells to apoptosis induction, suggesting that proteasomal inhibition may be a promising combination strategy for glioma therapeutics. In this study, we examined whether bortezomib could enhance response to HDAC inhibition in glioma cells. Although primary cells from glioblastoma multiforme (GBM) patients and established glioma cell lines did not show significant induction of apoptosis with vorinostat treatment alone, the combination of vorinostat plus bortezomib significantly enhanced apoptosis. The enhanced efficacy was due to proapoptotic mitochondrial injury and increased generation of reactive oxygen species. Our results also revealed that combination of bortezomib with vorinostat enhanced apoptosis by increasing Mcl-1 cleavage, Noxa upregulation, Bak and Bax activation, and cytochrome c release. Further downregulation of Mcl-1 using shRNA enhanced cell killing by the bortezomib/vorinostat combination. Vorinostat induced a rapid and sustained phosphorylation of histone H2AX in primary GBM and T98G cells, and this effect was significantly enhanced by co-administration of bortezomib. Vorinostat/bortezomib combination also induced Rad51 downregulation, which plays an important role in the synergistic enhancement of DNA damage and apoptosis. The significantly enhanced antitumor activity that results from the combination of bortezomib and HDACIs offers promise as a novel treatment for glioma patients.


Subject(s)
Apoptosis/drug effects , Boronic Acids/pharmacology , Central Nervous System Neoplasms/drug therapy , Glioma/drug therapy , Glioma/genetics , Hydroxamic Acids/pharmacology , Pyrazines/pharmacology , Reactive Oxygen Species/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Boronic Acids/administration & dosage , Bortezomib , Cell Line, Tumor , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Cytochromes c/metabolism , DNA Damage/drug effects , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioma/metabolism , Glioma/pathology , Histones/metabolism , Humans , Hydroxamic Acids/administration & dosage , Membrane Potential, Mitochondrial/drug effects , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation , Proteasome Inhibitors/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrazines/administration & dosage , Tumor Cells, Cultured , Vorinostat , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
3.
J Pharmacol Exp Ther ; 341(3): 859-72, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22393246

ABSTRACT

We observed that glioma cells are differentially sensitive to N-{4-[4-(4'-chloro-biphenyl-2-ylmethyl)-piperazin-1-yl]-benzoyl}-4-(3-dimethylamino-1-phenylsulfanylmethyl-propylamino)-3-nitro-benzenesulfonamide (ABT-737) and administration of ABT-737 at clinically achievable doses failed to induce apoptosis. Although elevated Bcl-2 levels directly correlated with sensitivity to ABT-737, overexpression of Bcl-2 did not influence sensitivity to ABT-737. To understand the molecular basis for variable and relatively modest sensitivity to the Bcl-2 homology domain 3 mimetic drug ABT-737, the abundance of Bcl-2 family members was assayed in a panel of glioma cell lines. Bcl-2 family member proteins, Bcl-xL, Bcl-w, Mcl-1, Bax, Bak, Bid, and Noxa, were found to be expressed ubiquitously at similar levels in all cell lines tested. We then examined the contribution of other apoptosis-resistance pathways to ABT-737 resistance. Bortezomib, an inhibitor of nuclear factor-kappaB (NF-κB), was found to enhance sensitivity of ABT-737 in phosphatase and tensin homolog on chromosome 10 (PTEN)-wild type, but not PTEN-mutated glioma cell lines. We therefore investigated the association between phosphatidylinositol 3-kinase (PI3K)/Akt activation and resistance to the combination of ABT-737 and bortezomib in PTEN-deficient glioma cells. Genetic and pharmacological inhibition of PI3K inhibition sensitized PTEN-deficient glioma cells to bortezomib- and ABT-737-induced apoptosis by increasing cleavage of Bid protein, activation and oligomerization of Bax, and loss of mitochondrial membrane potential. Our data further suggested that PI3K/Akt-dependent protection may occur upstream of the mitochondria. This study demonstrates that interference with multiple apoptosis-resistance signaling nodes, including NF-κB, Akt, and Bcl-2, may be required to induce apoptosis in highly resistant glioma cells, and therapeutic strategies that target the PI3K/Akt pathway may have a selective role for cancers lacking PTEN function.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , Biphenyl Compounds/pharmacology , Boronic Acids/pharmacology , Mitochondrial Diseases/metabolism , Nitrophenols/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyrazines/pharmacology , Sulfonamides/pharmacology , bcl-2-Associated X Protein/metabolism , Annexins/metabolism , Blotting, Western , Bortezomib , Cell Line, Tumor/metabolism , Cell Proliferation , Drug Synergism , Glioma/metabolism , Glioma/pathology , Humans , NF-kappa B/metabolism , Piperazines/pharmacology
4.
J Carcinog ; 92010 Jul 14.
Article in English | MEDLINE | ID: mdl-20808823

ABSTRACT

BACKGROUND: Src family kinases (SFK) collectively regulate a variety of cellular functions in many cancer types, including proliferation, invasion, motility, survival, differentiation, and angiogenesis. Although Dasatinib (BMS-354825), an ATP-competitive, small molecule tyrosine kinase inhibitor, suppresses the activity of SFKs at nanomolar concentrations, IC50 values for antiproliferative effects in glioma cell lines were well above the clinically achievable range, suggesting the need to interfere with other components of receptor-induced downstream signaling in order to achieve an optimal therapeutic effect. MATERIALS AND METHODS: The cytotoxic effects of combining Src and STAT3 inhibition on glioma cell lines were evaluated using assays to measure cell proliferation, apoptosis and migration. Western blotting and immunocytochemistry was used to monitor its effects on cell signaling and morphology. RESULTS: Silencing Src and STAT3 expression each partially inhibited cell proliferation and migration. In addition, JSI-124 significantly enhanced the efficacy of dasatinib in vitro. Combination of dasatinib and JSI-124 achieved significant inhibition of migration in all cell lines, which correlated with the inhibition of Src and downstream mediators of adhesion (e.g. focal adhesion kinase). Cells exposed to dasatinib and JSI-124 exhibited morphological changes that were consistent with an upstream role for Src in regulating focal adhesion complexes. CONCLUSIONS: Targeting the Src and STAT pathways may contribute to the treatment of cancers that demonstrate increased levels of these signaling mediators, including malignant human glioma. Clinical studies in these tumor types are warranted.

5.
Int J Cancer ; 126(10): 2282-95, 2010 May 15.
Article in English | MEDLINE | ID: mdl-19795461

ABSTRACT

Glioblastomas, the most malignant type of glioma, are more glycolytic than normal brain tissue. Robust migration of glioblastoma cells has been previously demonstrated under glycolytic conditions and their pseudopodia contain increased glycolytic and decreased mitochondrial enzymes. Glycolysis is suppressed by metabolic acids, including citric acid which is excluded from mitochondria during hypoxia. We postulated that glioma cells maintain glycolysis by regulating metabolic acids, especially in their pseudopodia. The enzyme that breaks down cytosolic citric acid is ATP citrate lyase (ACLY). Our identification of increased ACLY in pseudopodia of U87 glioblastoma cells on 1D gels and immunoblots prompted investigation of ACLY gene expression in gliomas for survival data and correlation with expression of ENO1, that encodes enolase 1. Queries of the NIH's REMBRANDT brain tumor database based on Affymetrix data indicated that decreased survival correlated with increased gene expression of ACLY in gliomas. Queries of gliomas and glioblastomas found an association of upregulated ACLY and ENO1 expression by chi square for all probe sets (reporters) combined and correlation for numbers of probe sets indicating shared upregulation of these genes. Real-time quantitative PCR confirmed correlation between ACLY and ENO1 in 21 glioblastomas (p < 0.001). Inhibition of ACLY with hydroxycitrate suppressed (p < 0.05) in vitro glioblastoma cell migration, clonogenicity and brain invasion under glycolytic conditions and enhanced the suppressive effects of a Met inhibitor on cell migration. In summary, gene expression data, proteomics and functional assays support ACLY as a positive regulator of glycolysis in glioblastomas.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Glioma/metabolism , Glycolysis , Phosphopyruvate Hydratase/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Brain Neoplasms/enzymology , Cell Line, Tumor , Cell Movement/drug effects , Chi-Square Distribution , Citrates/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioma/enzymology , Humans , Macrolides/pharmacology , Polymerase Chain Reaction , Protein-Tyrosine Kinases/antagonists & inhibitors , Pseudopodia/enzymology , Rats , Up-Regulation
6.
Cancer Lett ; 255(1): 135-44, 2007 Sep 18.
Article in English | MEDLINE | ID: mdl-17543444

ABSTRACT

Studies of primary cells from malignant brain tumors such as glioblastomas are limited by the small size of surgically resected specimens. However, glioblastomas are also frequently debulked via ultrasonic aspiration. In this study, we examined the functional competence and growth of their aspirated cells. Cells from minced tissue and aspirations were comparable in migration, formation of pseudopodia, development of cellular spheres with radial outgrowth, and neuroectodermal features. Cultures were maintained for more than six weeks without fibroblastic overgrowth. Our observations show that ultrasonically aspirated specimens contain cells useful for studies of tumor migration and growth of tumorspheres.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Brain/pathology , Cell Communication , Cell Movement , Cell Proliferation , Culture Media, Conditioned/pharmacology , Fibroblasts/metabolism , Humans , Microscopy, Fluorescence/methods , Neoplasm Invasiveness , Pseudopodia/pathology , Tumor Cells, Cultured , Ultrasonography
7.
Lab Invest ; 86(11): 1103-14, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16969371

ABSTRACT

It is well accepted that dysfunction in the blood brain barrier (BBB) allows permeation of albumin from the bloodstream into astrocytic brain tumors, especially glioblastomas, the most aggressive astrocytomas. In vitro, bovine serum albumin (BSA) aids functional cell assays by maintaining cytokines and growth factors in solution and delivering its cargo of fatty acids. Earlier, we showed that BSA was prominent in lysates prepared from pseudopodia formed by U87 astrocytoma cells. The present studies investigated the association of albumin with pseudopodia formed by U87 and LN229 astrocytoma cells. With hepatocyte growth factor (HGF) stimulation, cell migration was enhanced and BSA, especially its dimerized form, was prominent in pseudopodia compared to unmigrated cells on one-dimensional gels and immunoblots. When lysates were equalized for levels of glyceraldehyde-3-phosphate dehydrogenase, the rise for BSA levels in pseudopodia vs migrated cells was comparable or greater than levels noted for established pseudopodial proteins, beta-actin and ezrin. The increase for dimerized BSA in pseudopodia compared to unmigrated cells was greater than the rise in levels of beta-actin, ezrin, HGF, and phosphorylated Met when pseudopodia were harvested from filters with 1 mum pores using either cell line. Fluorescein (F)-labeled BSA co-localized with HGF on actin-rich cellular protrusions and with CM-DiI labeled pseudopodial plasma membranes. The F-BSA highlighted small, individual pseudopodial profiles more so than complex pseudopodial networks (reticulopodia) or unmigrated cells. Labeled human serum albumin also decorated pseudopodia preferentially. Albumin's association with pseudopodia may help to explain its selective accumulation in astrocytomas in vivo. The leaky BBB permits serum albumin to enter the microenvironment of astrocytomas thus allowing their invasive cells contact with serum albumin as a source of fatty acids that would be useful for remodeling cell membranes in pseudopodia. Thus, albumin potentially aids and marks invasion as it accumulates in these tumors.


Subject(s)
Astrocytoma/drug therapy , Hepatocyte Growth Factor/pharmacology , Pseudopodia/metabolism , Serum Albumin, Bovine/metabolism , Serum/physiology , Actins/pharmacology , Animals , Astrocytoma/pathology , Cattle , Cell Line, Tumor/drug effects , Cell Line, Tumor/pathology , Cell Movement/drug effects , Cytoskeletal Proteins/pharmacology , Humans , Immunoblotting , Microscopy, Fluorescence , Pseudopodia/drug effects , Pseudopodia/pathology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
8.
Lab Invest ; 85(12): 1457-70, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16170333

ABSTRACT

Increased glycolysis is characteristic of malignancy. Previously, with a mitochondrial inhibitor, we demonstrated that glycolytic ATP production was sufficient to support migration of melanoma cells. Recently, we found that glycolytic enzymes were abundant and some were increased in pseudopodia formed by U87 glioma (astrocytoma) cells. In this study, we examined cell migration, adhesion (a step in migration), and Matrigel invasion of U87 and LN229 glioma cells when their mitochondria were inhibited with sodium azide or limited by 1% O(2). Cell migration, adhesion, and invasion were comparable, with and without mitochondrial inhibition. Upon discovering that glycolysis alone can support glioma cell migration, unique features of glucose metabolism in astrocytic cells were investigated. The ability of astrocytic cells to remove lactate, the inhibitor of glycolysis, via gluconeogenesis and incorporation into glycogen led to consideration of supportive genetic mutations. Loss of phosphatase and tensin homolog (PTEN) releases glycogenesis from constitutive inhibition by glycogen synthase kinase-3 (GSK3). We hypothesize that glycolysis in gliomas can support invasive migration, especially when aided by loss of PTEN's regulation on the phosphatidylinositol-3 kinase (PI3K)/Akt pathway leading to inhibition of GSK3. Migration of PTEN-mutated U87 cells was studied for release of extracellular lactic acid and support by gluconeogenesis, loss of PTEN, and active PI3K. Lactic acid levels plateaued and phosphorylation changes confirmed activation of the PI3K/Akt pathway and glycogen synthase when cells relied only on glycolysis. Glycolytic U87 cell migration and phosphorylation of GSK3 were inhibited by PTEN transfection. Glycolytic migration was also suppressed by inhibiting PI3K and gluconeogenesis with wortmannin and metformin, respectively. These findings confirm that glycolytic glioma cells can migrate invasively and that the loss of PTEN is supportive, with activated glycogenic potential included among the relevant downstream effects.


Subject(s)
Astrocytoma/metabolism , Enzyme Inhibitors/pharmacology , Gluconeogenesis/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycolysis/physiology , PTEN Phosphohydrolase/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Astrocytoma/genetics , Astrocytoma/pathology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Collagen , Drug Combinations , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Humans , Lactic Acid/metabolism , Laminin , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasm Invasiveness/physiopathology , PTEN Phosphohydrolase/genetics , Phosphorylation , Proteoglycans , RNA, Messenger/metabolism , Sodium Azide/pharmacology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...