Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Cancer ; 124(1): 55-67, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18821579

ABSTRACT

Malignant rhabdoid tumors (MRT) exhibit a very poor prognosis because of their resistance to chemotherapeutic agents and new therapies are needed for the treatment of this cancer. Here, we show that the histone deacetylase (HDAC) inhibitor FK228 (depsipeptide) has an antitumor effect on MRT cells both in vitro and in vivo. FK228 is a unique cyclic peptide and is among the most potent inhibitors of both Class I and Class II HDACs. FK228 inhibited proliferation and induced apoptosis in all MRT cell lines tested. Preincubation with the pancaspase inhibitor zVAD-fmk did not completely rescue FK228-induced cell death, although it did inhibit apoptosis. Transmission electron microscopy (TEM) showed that FK228 could stimulate MRT cells to undergo apoptosis, necrosis or autophagy. FK228 converted unconjugated microtubule-associated protein light chain 3 (LC3-I) to conjugated light chain 3 (LC3-II) and induced localization of LC3 to autophagosomes. Apoptosis inducing factor (AIF), which plays a role in caspase-independent cell death, translocated to the nucleus in response to FK228 treatment. Moreover, small interfering RNA (siRNA) targeting of AIF prevented the morphological changes associated with autophagy and redistribution of LC3 to autophagosomes. Disrupting autophagy with chloroquine treatment enhanced FK228-induced cell death. In vivo, FK228 caused a reduction in tumor size and induced autophagy in tumor tissues. Using immunoelectron microscopy, we confirmed AIF translocation into the nucleus of FK228-induced autophagic cells in vivo. Thus, FK228 is a novel candidate for an antitumor agent for MRT cells.


Subject(s)
Apoptosis Inducing Factor/metabolism , Autophagy , Depsipeptides/pharmacology , Enzyme Inhibitors/pharmacology , Rhabdoid Tumor/drug therapy , Rhabdoid Tumor/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Nucleus/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Prognosis , Protein Transport , Rhabdoid Tumor/pathology
2.
Eur J Pharmacol ; 516(3): 187-96, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15963976

ABSTRACT

We have previously reported that calcium ionophore A23187 differentially induces necrosis in CEM cells, a T-lymphoblastic leukemia cell line, and apoptosis in HL60 cells, a promyelocytic leukemia cell line. Stimulation with VP16, however, induces typical apoptosis in both cell lines. Necrosis in CEM cells, characterized by cell shrinkage and clustering, began within 5 min of treatment. Swelling of the mitochondria, lumpy chromatin condensation and intact plasma membranes were evident by electron microscopy. These A23187-mediated changes in CEM cells were suppressed by clonazepam or CGP37157, inhibitors of the mitochondrial Na(+)/Ca(2+) exchanger. The changes, however, were not affected by cyclosporin A, an inhibitor of the mitochondrial permeability transition pore. In both CEM and HL60 cells, intra-cellular calcium increased with similar amplitude within 1 min of treatment with 2 microM A23187. Intra-mitochondrial calcium increased with clonazepam pre-treatment alone in both CEM and HL60 cells. However, intra-mitochondrial calcium did not change drastically in response to A23187 in CEM or HL60 cells, either untreated or pre-treated with clonazepam. A23187 induces necrosis in CEM cells concurrent with mitochondrial dysfunction, which is independent of the mitochondrial permeability transition, but affected by intra-mitochondrial calcium, while HL60 cells lack these early changes. Differences in the responses to A23187 between these two cell lines might derive from differences in the susceptibility of the mitochondrial membrane to rapid increases in intra-cellular calcium.


Subject(s)
Apoptosis/drug effects , Calcimycin/pharmacology , Mitochondria/drug effects , Calcium/metabolism , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , Cell Shape/drug effects , Clonazepam/analogs & derivatives , Clonazepam/pharmacology , Cyclosporine/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Flow Cytometry , HL-60 Cells , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Ionophores/pharmacology , Membrane Potentials/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Necrosis , Phospholipases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Thiazepines/pharmacology , Time Factors
3.
Blood ; 103(6): 2299-307, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-14645012

ABSTRACT

Caspase-independent programmed cell death can exhibit either an apoptosis-like or a necrosis-like morphology. The ABL kinase inhibitor, imatinib mesylate, has been reported to induce apoptosis of BCR-ABL-positive cells in a caspase-dependent fashion. We investigated whether caspases alone were the mediators of imatinib mesylate-induced cell death. In contrast to previous reports, we found that a broad caspase inhibitor, zVAD-fmk, failed to prevent the death of imatinib mesylate-treated BCR-ABL-positive human leukemic cells. Moreover, zVAD-fmk-preincubated, imatinib mesylate-treated cells exhibited a necrosis-like morphology characterized by cellular pyknosis, cytoplasmic vacuolization, and the absence of nuclear signs of apoptosis. These cells manifested a loss of the mitochondrial transmembrane potential, indicating the mitochondrial involvement in this caspase-independent necrosis. We excluded the participation of several mitochondrial factors possibly involved in caspase-independent cell death such as apoptosis-inducing factor, endonuclease G, and reactive oxygen species. However, we observed the mitochondrial release of the serine protease Omi/HtrA2 into the cytosol of the cells treated with imatinib mesylate or zVAD-fmk plus imatinib mesylate. Furthermore, serine protease inhibitors prevented the caspase-independent necrosis. Taken together, our results suggest that imatinib mesylate induces a caspase-independent, necrosis-like programmed cell death mediated by the serine protease activity of Omi/HtrA2.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Piperazines/pharmacology , Pyrimidines/pharmacology , Serine Endopeptidases/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/physiology , Apoptosis Inducing Factor , Benzamides , Caspase 3 , Caspase 9 , Caspase Inhibitors , Caspases/metabolism , Cell Line, Tumor/cytology , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Cell Nucleus/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Flavoproteins/metabolism , Fusion Proteins, bcr-abl/metabolism , High-Temperature Requirement A Serine Peptidase 2 , Humans , Imatinib Mesylate , Membrane Potentials/physiology , Membrane Proteins/metabolism , Mitochondria/physiology , Mitochondrial Proteins , Necrosis , Nucleosomes/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...