Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 337(1): 65-74, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21228063

ABSTRACT

Exposure of the human malignant peripheral nerve sheath tumor cell lines STS-26T, ST88-14, and NF90-8 to nanomolar concentrations of both lovastatin and farnesyl transferase inhibitor (FTI)-1 but not to either drug alone induced cell death. ST88-14 and NF90-8 cells underwent apoptosis, yet dying STS-26T cells did not. FTI-1 cotreatment induced a strong and sustained autophagic response as indicated by analyses of microtubule-associated protein-1 light chain 3 (LC3)-II accumulation in STS-26T cultures. Extensive colocalization of LC3-positive punctate spots was observed with both lysosome-associated membrane protein (LAMP)-1 and LAMP-2 (markers of late endosomes/lysosomes) in solvent or FTI-1 or lovastatin-treated STS-26T cultures but very little colocalization in lovastatin/FTI-1-cotreated cultures. The absence of colocalization in the cotreatment protocol correlated with loss of LAMP-2 expression. Autophagic flux studies indicated that lovastatin/FTI-1 cotreatment inhibited the completion of the autophagic program. In contrast, rapamycin induced an autophagic response that was associated with cytostasis but maintenance of viability. These studies indicate that cotreatment of STS-26T cells with lovastatin and FTI-1 induces an abortive autophagic program and nonapoptotic cell death.


Subject(s)
Apoptosis , Autophagy/drug effects , Enzyme Inhibitors/administration & dosage , Farnesyltranstransferase/antagonists & inhibitors , Lovastatin/administration & dosage , Animals , Autophagy/physiology , Cell Death/drug effects , Cell Death/physiology , Cell Line , Cell Line, Tumor , Cells, Cultured , Drug Combinations , Farnesyltranstransferase/metabolism , Humans , Mice , Rats
2.
Free Radic Biol Med ; 50(2): 281-94, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21074609

ABSTRACT

Reactive oxygen species (ROS) can induce lysosomal membrane permeabilization (LMP). Photoirradiation of murine hepatoma 1c1c7 cultures preloaded with the photosensitizer NPe6 generates singlet oxygen within acidic organelles and causes LMP and the activation of procaspases. Treatment with the cationic amphiphilic drugs (CADs) U18666A, imipramine, and clozapine stimulated the accumulation of filipin-stainable nonesterified cholesterol/sterols in late endosomes/lysosomes, but not in mitochondria. Concentration-response studies demonstrated an inverse relationship between lysosomal nonesterified cholesterol/sterol contents and susceptibility to NPe6 photoirradiation-induced intracellular membrane oxidation, LMP, and activation of procaspase-9 and -3. Similarly, the kinetics of restoration of NPe6 photoirradiation-induced LMP paralleled the losses of lysosomal cholesterol that occurred upon replating U18666A-treated cultures in CAD-free medium. Consistent with the oxidation of lysosomal cholesterol, filipin staining in U18666A-treated cultures progressively decreased with increasing photoirradiating light dose. U18666A also suppressed the induction of LMP and procaspase activation by exogenously added hydrogen peroxide. However, neither U18666A nor imipramine suppressed the induction of apoptosis by agents that did not directly induce LMP. These studies indicate that lysosomal nonesterified cholesterol/sterol content modulates susceptibility to ROS-induced LMP and possibly does so by being an alternative target for oxidants and lowering the probability of damage to other lysosomal membrane lipids and/or proteins.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Membrane Permeability , Cholesterol/metabolism , Intracellular Membranes/metabolism , Liver Neoplasms/metabolism , Lysosomes/metabolism , Oxidants/pharmacology , Androstenes/pharmacology , Animals , Anticholesteremic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Hepatocellular/pathology , Caspase 9/metabolism , Cholesterol/chemistry , Endosomes/drug effects , Endosomes/metabolism , Endosomes/radiation effects , Filipin/metabolism , Intracellular Membranes/drug effects , Intracellular Membranes/radiation effects , Light , Liver Neoplasms/pathology , Lysosomes/drug effects , Lysosomes/radiation effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/radiation effects , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
3.
J Pharmacol Exp Ther ; 328(3): 796-806, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19066341

ABSTRACT

The therapeutic usefulness of the quinoxaline derivatives XK469 (2-{4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy}propionic acid) and SH80 (2-{4-[(7-bromo-2-quinolinyl)oxy]phenoxy}propionic acid) has been attributed to their abilities to induce G(2)/M arrest and apoptotic or autophagic cell death. Concentrations of XK469 or SH80 > or = 5 microM were cytostatic to cultures of the normal murine melanocyte cell line Melan-a. Higher concentrations caused dose-dependent cytotoxicity. Concentrations > or =10 microM provoked dramatic morphological changes typified by marked increases in cell size and granularity. XK469/SH80-treated cultures accumulated tetraploid (4N) DNA-containing cells within 24 h of treatment, an 8N population within 3 days, and a 16N population within 5 days. Increases in ploidy correlated with the appearance of multinucleated cells. Under no circumstances did cells exhibit evidence of furrow formation. Both drugs suppressed cytokinesis in additional mammalian cell lines. Cytotoxic concentrations of XK469 elevated DEVDase activities (a measure of procaspase-3/7 activation) and enhanced cellular staining by a fluorescent analog of the pan caspase inhibitor valine-alanine-aspartic acid-fluoromethyl ketone within 48 to 96 h of treatment. Within 48 h of treatment, cytostatic and cytotoxic concentrations of XK469 elevated p21 contents, reduced Bcl-2 and Bcl-XL contents, and induced autophagy, as monitored by the accumulation of phosphatidylethanolamine-modified cleavage product of microtubule-associated protein light chain 3 (LC3-II). Cultures treated with > or =10 microM XK469 or SH80 for 5 days could not be induced to divide upon removal of drugs. Such cultures maintained high LC3-II contents, exhibited reduced cyclin E and D1 contents, and extensively expressed senescence-associated beta-galactosidase within 14 to 17 days of cessation of drug treatment. Hence, XK469 and SH80 inhibit cytokinesis, promote polyploidy, and induce senescence in Melan-a cells.


Subject(s)
Cellular Senescence/drug effects , Cytokinesis/drug effects , Polyploidy , Propionates/pharmacology , Quinoxalines/pharmacology , Animals , Autophagy/drug effects , Cell Division/drug effects , Cell Line/drug effects , Cells, Cultured , Flow Cytometry , Mice , Peptide Hydrolases/metabolism
4.
J Pharmacol Exp Ther ; 326(1): 1-11, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18367665

ABSTRACT

Neurofibromatosis type 1 (NF1) is a genetic disorder that is driven by the loss of neurofibromin (Nf) protein function. Nf contains a Ras-GTPase-activating protein domain, which directly regulates Ras signaling. Numerous clinical manifestations are associated with the loss of Nf and increased Ras activity. Ras proteins must be prenylated to traffic and functionally localize with target membranes. Hence, Ras is a potential therapeutic target for treating NF1. We have tested the efficacy of two novel farnesyl transferase inhibitors (FTIs), 1 and 2, alone or in combination with lovastatin, on two NF1 malignant peripheral nerve sheath tumor (MPNST) cell lines, NF90-8 and ST88-14. Single treatments of 1, 2, or lovastatin had no effect on Ras prenylation or MPNST cell proliferation. However, low micromolar combinations of 1 or 2 with lovastatin (FTI/lovastatin) reduced Ras prenylation in both MPNST cell lines. Furthermore, this FTI/lovastatin combination treatment reduced cell proliferation and induced an apoptotic response as shown by morphological analysis, procaspase-3/-7 activation, loss of mitochondrial membrane potential, and accumulation of cells with sub-G(1) DNA content. Little to no detectable toxicity was observed in normal rat Schwann cells following FTI/lovastatin combination treatment. These data support the hypothesis that combination FTI plus lovastatin therapy may be a potential treatment for NF1 MPNSTs.


Subject(s)
Apoptosis/drug effects , Farnesyltranstransferase/antagonists & inhibitors , Lovastatin/administration & dosage , Nerve Sheath Neoplasms/drug therapy , Nerve Sheath Neoplasms/pathology , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/pathology , Animals , Animals, Newborn , Apoptosis/physiology , Cell Line , Cell Line, Transformed , Cell Line, Tumor , Drug Therapy, Combination , Enzyme Inhibitors/administration & dosage , Farnesyltranstransferase/metabolism , Lovastatin/chemistry , Neurofibromatosis 1/enzymology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...