Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cell Rep ; 27(3): 820-834.e9, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995479

ABSTRACT

Inhibition of oxidative phosphorylation (OXPHOS) by 1-cyclopropyl-4-(4-[(5-methyl-3-(3-[4-(trifluoromethoxy)phenyl]-1,2,4-oxadiazol-5-yl)-1H-pyrazol-1-yl)methyl]pyridin-2-yl)piperazine (BAY87-2243, abbreviated as B87), a complex I inhibitor, fails to kill human cancer cells in vitro. Driven by this consideration, we attempted to identify agents that engage in synthetically lethal interactions with B87. Here, we report that dimethyl α-ketoglutarate (DMKG), a cell-permeable precursor of α-ketoglutarate that lacks toxicity on its own, kills cancer cells when combined with B87 or other inhibitors of OXPHOS. DMKG improved the antineoplastic effect of B87, both in vitro and in vivo. This combination caused MDM2-dependent, tumor suppressor protein p53 (TP53)-independent transcriptional reprogramming and alternative exon usage affecting multiple glycolytic enzymes, completely blocking glycolysis. Simultaneous inhibition of OXPHOS and glycolysis provoked a bioenergetic catastrophe culminating in the activation of a cell death program that involved disruption of the mitochondrial network and activation of PARP1, AIFM1, and APEX1. These results unveil a metabolic liability of human cancer cells that may be harnessed for the development of therapeutic regimens.


Subject(s)
Apoptosis/drug effects , Electron Transport Complex I/antagonists & inhibitors , Ketoglutaric Acids/pharmacology , Animals , Apoptosis Inducing Factor/metabolism , Cell Line, Tumor , Electron Transport Complex I/metabolism , Female , Glycolysis/drug effects , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mice , Mice, Nude , Mitochondria/metabolism , Oxadiazoles/pharmacology , Oxidative Phosphorylation/drug effects , Poly (ADP-Ribose) Polymerase-1/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/metabolism , Pyrazoles/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism
2.
Cell Death Dis ; 10(4): 301, 2019 Apr 03.
Article in English | MEDLINE | ID: mdl-30944306

ABSTRACT

In the version of this article originally submitted, it was stated that the first three authors (Shaoyi_ Than, Yan Wang, Wei Xie) had contributed equally. However, in the published version this information was missing.

3.
Cell Death Dis ; 10(3): 242, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30858361

ABSTRACT

Cystic fibrosis is a disease caused by defective function of a chloride channel coupled to a blockade of autophagic flux. It has been proposed to use synthetic chloride transporters as pharmacological agents to compensate insufficient chloride fluxes. Here, we report that such chloride anionophores block autophagic flux in spite of the fact that they activate the pro-autophagic transcription factor EB (TFEB) coupled to the inhibition of the autophagy-suppressive mTORC1 kinase activity. Two synthetic chloride transporters (SQ1 and SQ2) caused a partially TFEB-dependent relocation of the autophagic marker LC3 to the Golgi apparatus. Inhibition of TFEB activation using a calcium chelator or calcineurin inhibitors reduced the formation of LC3 puncta in cells, yet did not affect the cytotoxic action of SQ1 and SQ2 that could be observed after prolonged incubation. In conclusion, the squaramide-based synthetic chloride transporters studied in this work (which can also dissipate pH gradients) are probably not appropriate for the treatment of cystic fibrosis yet might be used for other indications such as cancer.


Subject(s)
Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Hydrocarbons, Fluorinated/pharmacology , Ion Transport/drug effects , TOR Serine-Threonine Kinases/metabolism , Autophagy/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Death , Cell Line, Tumor , Golgi Apparatus/drug effects , Humans , Hydrocarbons, Fluorinated/chemistry , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/genetics , Up-Regulation
4.
Cell Death Dis ; 9(2): 191, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29415993

ABSTRACT

Cystic Fibrosis (CF) due to the ΔF508 mutation of cystic fibrosis transmembrane conductance regulator (CFTR) can be treated with a combination of cysteamine and Epigallocatechin gallate (EGCG). Since ECGC is not a clinically approved drug, we attempted to identify other compounds that might favourably interact with cysteamine to induce autophagy and thus rescuing the function of ΔF508 CFTR as a chloride channel in the plasma membrane. For this, we screened a compound library composed by chemically diverse autophagy inducers for their ability to enhance autophagic flux in the presence of cysteamine. We identified the antiarrhythmic Ca2+ channel blocker amiodarone, as an FDA-approved drug having the property to cooperate with cysteamine to stimulate autophagy in an additive manner. Amiodarone promoted the re-expression of ΔF508 CFTR protein in the plasma membrane of respiratory epithelial cells. Hence, amiodarone might be yet another compound for the etiological therapy of CF in patients bearing the ΔF508 CFTR mutation.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Cystic Fibrosis/pathology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Autophagy/drug effects , Bronchi/cytology , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Line , Cell Line, Tumor , Cysteamine/pharmacology , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Humans , Transfection
5.
Mol Cell ; 53(5): 710-25, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24560926

ABSTRACT

Acetyl-coenzyme A (AcCoA) is a major integrator of the nutritional status at the crossroads of fat, sugar, and protein catabolism. Here we show that nutrient starvation causes rapid depletion of AcCoA. AcCoA depletion entailed the commensurate reduction in the overall acetylation of cytoplasmic proteins, as well as the induction of autophagy, a homeostatic process of self-digestion. Multiple distinct manipulations designed to increase or reduce cytosolic AcCoA led to the suppression or induction of autophagy, respectively, both in cultured human cells and in mice. Moreover, maintenance of high AcCoA levels inhibited maladaptive autophagy in a model of cardiac pressure overload. Depletion of AcCoA reduced the activity of the acetyltransferase EP300, and EP300 was required for the suppression of autophagy by high AcCoA levels. Altogether, our results indicate that cytosolic AcCoA functions as a central metabolic regulator of autophagy, thus delineating AcCoA-centered pharmacological strategies that allow for the therapeutic manipulation of autophagy.


Subject(s)
Acetyl Coenzyme A/chemistry , Autophagy , Cytosol/enzymology , Gene Expression Regulation, Enzymologic , Adenosine Triphosphate/chemistry , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Cytosol/metabolism , E1A-Associated p300 Protein/chemistry , Green Fluorescent Proteins/metabolism , HCT116 Cells , HeLa Cells , Humans , Ketoglutaric Acids/chemistry , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Mitochondria/metabolism , RNA, Small Interfering/metabolism
6.
Cell Cycle ; 11(20): 3851-60, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-23070521

ABSTRACT

Resveratrol is a polyphenol contained in red wine that has been amply investigated for its beneficial effects on organismal metabolism, in particular in the context of the so-called "French paradox," i.e., the relatively low incidence of coronary heart disease exhibited by a population with a high dietary intake of cholesterol and saturated fats. At least part of the beneficial effect of resveratrol on human health stems from its capacity to promote autophagy by activating the NAD-dependent deacetylase sirtuin 1. However, the concentration of resveratrol found in red wine is excessively low to account alone for the French paradox. Here, we investigated the possibility that other mono- and polyphenols contained in red wine might induce autophagy while affecting the acetylation levels of cellular proteins. Phenolic compounds found in red wine, including anthocyanins (oenin), stilbenoids (piceatannol), monophenols (caffeic acid, gallic acid) glucosides (delphinidin, kuronamin, peonidin) and flavonoids (catechin, epicatechin, quercetin, myricetin), were all capable of stimulating autophagy, although with dissimilar potencies. Importantly, a robust negative correlation could be established between autophagy induction and the acetylation levels of cytoplasmic proteins, as determined by a novel immunofluorescence staining protocol that allows for the exclusion of nuclear components from the analysis. Inhibition of sirtuin 1 by both pharmacological and genetic means abolished protein deacetylation and autophagy as stimulated by resveratrol, but not by piceatannol, indicating that these compounds act through distinct molecular pathways. In support of this notion, resveratrol and piceatannol synergized in inducing autophagy as well as in promoting cytoplasmic protein deacetylation. Our results highlight a cause-effect relationship between the deacetylation of cytoplasmic proteins and autophagy induction by red wine components.


Subject(s)
Autophagy/drug effects , Cytoplasm/drug effects , Polyphenols/pharmacology , Sirtuin 1/genetics , Stilbenes/pharmacology , Acetylation , Autophagy/physiology , Cell Line, Tumor , Cytoplasm/enzymology , Enzyme Activation/drug effects , Gene Expression/drug effects , Humans , Microscopy, Fluorescence , Resveratrol , Sirtuin 1/metabolism
8.
Apoptosis ; 12(5): 803-13, 2007 May.
Article in English | MEDLINE | ID: mdl-17294081

ABSTRACT

Mitochondrial membrane permeabilization (MMP) is considered as the "point-of-no-return" in numerous models of programmed cell death. Indeed, mitochondria determine the intrinsic pathway of apoptosis, and play a major role in the extrinsic route as well. MMP affects the inner and outer mitochondrial membranes (IM and OM, respectively) to a variable degree. OM permeabilization culminates in the release of proteins that normally are confined in the mitochondrial intermembrane space (IMS), including caspase activators (e.g. cytochrome c) and caspase-independent death effectors (e.g. apoptosis-inducing factor). Partial IM permeabilization disrupts mitochondrial ion and volume homeostasis and dissipates the mitochondrial transmembrane potential (DeltaPsi(m)). The assessment of early mitochondrial alterations allows for the identification of cells that are committed to die but have not displayed yet the apoptotic phenotype. Several techniques to measure MMP by cytofluorometry and fluorescence microscopy have been developed. Here, we summarize the currently available methods for the detection of MMP, and provide a comparative analysis of these techniques.


Subject(s)
Apoptosis/physiology , Membrane Potentials/physiology , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Humans , Mitochondria/ultrastructure , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Permeability
10.
Biochim Biophys Acta ; 1659(2-3): 178-89, 2004 Dec 06.
Article in English | MEDLINE | ID: mdl-15576050

ABSTRACT

Mitochondrial membrane permeabilization (MMP) is a critical step regulating apoptosis. Viruses have evolved multiple strategies to modulate apoptosis for their own benefit. Thus, many viruses code for proteins that act on mitochondria and control apoptosis of infected cells. Viral proapoptotic proteins translocate to mitochondrial membranes and induce MMP, which is often accompanied by mitochondrial swelling and fragmentation. From a structural point of view, all the viral proapoptotic proteins discovered so far contain amphipathic alpha-helices that are necessary for the proapoptotic effects and seem to have pore-forming properties, as it has been shown for Vpr from human immunodeficiency virus-1 (HIV-1) and HBx from hepatitis B virus (HBV). In contrast, antiapoptotic viral proteins (e.g., M11L from myxoma virus, F1L from vaccinia virus and BHRF1 from Epstein-Barr virus) contain mitochondrial targeting sequences (MTS) in their C-terminus that are homologous to tail-anchoring domains. These domains are similar to those present in many proteins of the Bcl-2 family and are responsible for inserting the protein in the outer mitochondrial membrane leaving the N-terminus of the protein facing the cytosol. The antiapoptotic proteins K7 and K15 from avian encephalomyelitis virus (AEV) and viral mitochondria inhibitor of apoptosis (vMIA) from cytomegalovirus are capable of binding host-specific apoptosis-modulatory proteins such as Bax, Bcl-2, activated caspase 3, CAML, CIDE-B and HAX. In conclusion, viruses modulate apoptosis at the mitochondrial level by multiple different strategies.


Subject(s)
Apoptosis/physiology , DNA Viruses/metabolism , Mitochondria/metabolism , RNA Viruses/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Cell Death/physiology , Gene Products, vpr/metabolism , HIV-1/metabolism , Molecular Sequence Data , vpr Gene Products, Human Immunodeficiency Virus
11.
Proc Natl Acad Sci U S A ; 101(21): 7988-93, 2004 May 25.
Article in English | MEDLINE | ID: mdl-15148411

ABSTRACT

We report that the cytomegalovirus-encoded cell death suppressor vMIA binds Bax and prevents Bax-mediated mitochondrial membrane permeabilization by sequestering Bax at mitochondria in the form of a vMIA-Bax complex. vMIA mutants with a defective mitochondria-targeting domain retain their Bax-binding function but not their ability to suppress mitochondrial membrane permeabilization or cell death. vMIA does not seem to either specifically associate with Bak or suppress Bak-mediated mitochondrial membrane permeabilization. Recent evidence suggests that the contribution of Bax and Bak in the mitochondrial apoptotic signaling pathway depends on the distinct phenotypes of cells, and it appears from our data that vMIA is capable of suppressing apoptosis in cells in which this pathway is dominated by Bax, but not in cells where Bak also plays a role.


Subject(s)
Apoptosis , Cytomegalovirus/chemistry , Membrane Proteins/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Viral Proteins/metabolism , Animals , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/metabolism , Cell Line , Cell Membrane Permeability , Fibroblasts , HeLa Cells , Humans , Intracellular Membranes/metabolism , Membrane Proteins/genetics , Mice , Mitochondria/pathology , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Protein Transport , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Signal Transduction , Viral Proteins/chemistry , Viral Proteins/genetics , bcl-2 Homologous Antagonist-Killer Protein , bcl-2-Associated X Protein
12.
Methods Mol Biol ; 282: 103-15, 2004.
Article in English | MEDLINE | ID: mdl-15105559

ABSTRACT

Mitochondrial membrane permeabilization (MMP) constitutes an early event of the apoptotic process. MMP affects both mitochondrial membranes. Inner MMP leads to the dissipation of the inner transmembrane potential and outer MMP culminates in the efflux of apoptogenic factors. The exact molecular mechanisms of MMP are still controversial. A growing body of data suggests that the cell death regulatory activity of Bcl-2 family members depends, at least in some instances, on their ability to modulate the opening of the mitochondrial permeability transition pore complex. Here, we will detail some experimental protocols designed to measure mitochondrial membrane potential and permeability transition, either in intact cells or in isolated mitochondria.


Subject(s)
Apoptosis/physiology , Membrane Potentials , Mitochondria/metabolism , Animals , Cells, Cultured , Hepatocytes/cytology , Mice , Permeability , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats
13.
J Biol Chem ; 279(21): 22605-14, 2004 May 21.
Article in English | MEDLINE | ID: mdl-15004026

ABSTRACT

The viral mitochondria-localized inhibitor of apoptosis (vMIA), encoded by the UL37 gene of human cytomegalovirus, inhibits apoptosis-associated mitochondrial membrane permeabilization by a mechanism different from that of Bcl-2. Here we show that vMIA induces several changes in Bax that resemble those found in apoptotic cells yet take place in unstimulated, non-apoptotic vMIA-expressing cells. These changes include the constitutive localization of Bax at mitochondria, where it associates tightly with the mitochondrial membrane, forming high molecular weight aggregates that contain vMIA. vMIA recruits Bax to mitochondria but delays relocation of caspase-8-activated truncated Bid-green fluorescent protein (GFP) (t-Bid-GFP) to mitochondria. The ability of vMIA and its deletion mutants to associate with Bax and to induce relocation of Bax to mitochondria correlates with their anti-apoptotic activity and with their ability to suppress mitochondrial membrane permeabilization. Taken together, our data indicate that vMIA blocks apoptosis via its interaction with Bax. vMIA neutralizes Bax by recruiting it to mitochondria and "freezing" its pro-apoptotic activity. These data unravel a novel strategy of subverting an intrinsic pathway of apoptotic signaling.


Subject(s)
Apoptosis , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/physiology , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Viral Proteins/biosynthesis , Viral Proteins/metabolism , Viral Proteins/physiology , Animals , Caspase 8 , Caspases/metabolism , Cell Line , Cell-Free System , Cells, Cultured , Chromatography, Gel , Cytochromes c/metabolism , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence , Precipitin Tests , Transfection , bcl-2-Associated X Protein
14.
Biochem Pharmacol ; 66(8): 1321-9, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14555204

ABSTRACT

Cells expressing the envelope glycoprotein complex (Env) encoded by the human immunodeficiency virus can fuse with cells expressing Env receptors (CD4 and CXCR4). The resulting syncytia undergo apoptosis. We developed a cytofluorometric assay for the quantitation of syncytium formation and syncytial apoptosis. Using this methodology, we show that caspase activation in syncytia is inhibited by pharmacological or genetic intervention on cyclin-dependent kinase-1, p53, and mitochondrial membrane permeabilization (MMP). Thus, transfection of fusing cells with the viral mitochondrial inhibitor of apoptosis encoded by cytomegalovirus, a specific inhibitor of MMP, prevented the mitochondrial cytochrome c release and abolished simultaneously the activation of caspase-3. Conversely, inhibition of caspases did not prevent MMP. These results indicate that Env-elicited syncytial apoptosis involves the intrinsic (mitochondrial) pathway.


Subject(s)
Apoptosis , Caspases/metabolism , HIV-1/chemistry , Mitochondria/drug effects , Viral Envelope Proteins/pharmacology , CDC2 Protein Kinase/metabolism , Cytochromes c/metabolism , Enzyme Activation/drug effects , Flow Cytometry , Giant Cells/pathology , Giant Cells/virology , HeLa Cells , Humans , Mitochondria/enzymology
15.
J Neurochem ; 85(6): 1431-42, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12787063

ABSTRACT

Blockade of mitochondrial permeability transition protects against hypoglycemic brain damage. To study the mechanisms downstream from mitochondria that may cause neuronal death, we investigated the effects of cyclosporin A on subcellular localization of apoptosis-inducing factor and cytochrome c, activation of the cysteine proteases calpain and caspase-3, as well as its effect on brain extracellular calcium concentrations. Redistribution of cytochrome c occurred at 30 min of iso-electricity, whereas translocation of apoptosis-inducing factor to nuclei occurred at 30 min of recovery following 30 min of iso-electricity. Active caspase-3 and calpain-induced fodrin breakdown products were barely detectable in the dentate gyrus and CA1 region of the hippocampus of rat brain exposed to 30 or 60 min of insulin-induced hypoglycemia. However, 30 min or 3 h after recovery of blood glucose levels, fodrin breakdown products and active caspase-3 markedly increased, concomitant with a twofold increase in caspase-3-like enzymatic activity. When rats were treated with neuroprotective doses of cyclosporin A, but not with FK 506, the redistribution of apoptosis-inducing factor and cytochrome c was reduced and fodrin breakdown products and active caspase-3 immuno-reactivity was diminished whereas the extracellular calcium concentration was unaffected. We conclude that hypoglycemia leads to mitochondrial permeability transition which, upon recovery of energy metabolism, mediates the activation of caspase-3 and calpains, promoting cell death.


Subject(s)
Calcium/metabolism , Calpain/metabolism , Cyclosporine/pharmacology , Hypoglycemia/metabolism , Neurons/drug effects , Animals , Apoptosis Inducing Factor , Caspase 3 , Caspases/metabolism , Cell Count , Cytochrome c Group/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Enzyme Activation/drug effects , Flavoproteins/metabolism , Intracellular Fluid/metabolism , Male , Membrane Proteins/metabolism , Microelectrodes , Neurons/cytology , Neurons/metabolism , Protein Transport/drug effects , Rats , Rats, Wistar , Tacrolimus/pharmacology
16.
J Exp Med ; 197(10): 1323-34, 2003 May 19.
Article in English | MEDLINE | ID: mdl-12756268

ABSTRACT

A number of diseases are due to lysosomal destabilization, which results in damaging cell loss. To investigate the mechanisms of lysosomal cell death, we characterized the cytotoxic action of two widely used quinolone antibiotics: ciprofloxacin (CPX) or norfloxacin (NFX). CPX or NFX plus UV light (NFX*) induce lysosomal membrane permeabilization (LMP), as detected by the release of cathepsins from lysosomes. Inhibition of the lysosomal accumulation of CPX or NFX suppresses their capacity to induce LMP and to kill cells. CPX- or NFX-triggered LMP results in caspase-independent cell death, with hallmarks of apoptosis such as chromatin condensation and phosphatidylserine exposure on the plasma membrane. LMP triggers mitochondrial membrane permeabilization (MMP), as detected by the release of cytochrome c. Both CPX and NFX* cause Bax and Bak to adopt their apoptotic conformation and to insert into mitochondrial membranes. Bax-/- Bak-/- double knockout cells fail to undergo MMP and cell death in response to CPX- or NFX-induced LMP. The single knockout of Bax or Bak (but not Bid) or the transfection-enforced expression of mitochondrion-targeted (but not endoplasmic reticulum-targeted) Bcl-2 conferred protection against CPX (but not NFX*)-induced MMP and death. Altogether, our data indicate that mitochondria are indispensable for cell death initiated by lysosomal destabilization.


Subject(s)
Apoptosis , Cell Membrane Permeability , Lysosomes/metabolism , Mitochondria/physiology , Amino Acid Chloromethyl Ketones/pharmacology , Cell Membrane Permeability/drug effects , Ciprofloxacin/pharmacology , HeLa Cells , Humans , Membrane Proteins/physiology , Metalloendopeptidases/physiology , Norfloxacin/pharmacology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Reactive Oxygen Species , bcl-2 Homologous Antagonist-Killer Protein , bcl-2-Associated X Protein
17.
Biochem Biophys Res Commun ; 304(3): 575-81, 2003 May 09.
Article in English | MEDLINE | ID: mdl-12729592

ABSTRACT

During coevolution with their hosts, viruses have "learned" to intercept or to activate the principal signal transducing pathways leading to cell death. A number of proteins from pathophysiologically relevant viruses are targeted to mitochondria and regulate (induce or inhibit) the apoptosis-associated permeabilization of mitochondrial membranes. Such proteins are encoded by human immunodeficiency virus 1, Kaposi's sarcoma-associated herpesvirus, human T-cell leukemia virus-1, hepatitis B virus, cytomegalovirus, and Epstein Barr virus, among others. Within mitochondria, such apoptosis regulators from viral origin can target distinct proteins from the Bcl-2 family and the permeability transition pore complex including the adenine nucleotide translocase, cyclophilin D, the voltage-dependent anion channel, and the peripheral benzodiazepine receptor. Thus, viral proteins can regulate apoptosis at the mitochondrial level by acting on a variety of different targets.


Subject(s)
Apoptosis , Mitochondria/metabolism , Viral Proteins/pharmacology , Animals , Cell Membrane Permeability , Intracellular Membranes/metabolism , Mice , Mitochondrial Proteins/physiology
18.
J Leukoc Biol ; 73(3): 399-406, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12629154

ABSTRACT

We analyzed the mechanism of UVB-induced cell death using the Jurkat T cell line. Apoptosis was assessed by measuring phosphatidylserine (PS) externalization, caspase activity, the decrease in mitochondrial membrane potential (Delta Psi m), nucleosomal DNA fragmentation, and morphological changes such as chromatin condensation. The mitochondrio-nuclear translocation of apoptosis-inducing factor (AIF) was evaluated by confocal laser microscopy. The cell death pattern of UVB-irradiated cells was similar to the Fas-induced cell death pattern. However, zVAD-fmk inhibited the nucleosomal fragmentation of DNA but not the externalization of PS, decrease in Delta Psi m, or mitochondrio-nuclear translocation of AIF. N-acetyl L-cysteine significantly inhibited the translocation of AIF induced by UVB. These results suggested that caspase-dependent and -independent pathways were involved in UVB-induced cell death in Jurkat cells, and the mitochondrio-nuclear translocation of AIF was associated with the latter pathway. In addition, reactive oxygen species generated by UVB might be involved in inducing the mitochondrio-nuclear translocation of AIF.


Subject(s)
Cell Death/radiation effects , Flavoproteins/physiology , Membrane Proteins/physiology , Reactive Oxygen Species/radiation effects , T-Lymphocytes/radiation effects , Ultraviolet Rays , Apoptosis Inducing Factor , Caspases/metabolism , Cell Nucleus/metabolism , Flavoproteins/metabolism , Flavoproteins/radiation effects , Humans , Jurkat Cells , Membrane Proteins/metabolism , Membrane Proteins/radiation effects , Mitochondria/metabolism , Protein Transport/radiation effects , Reactive Oxygen Species/metabolism , T-Lymphocytes/cytology
19.
Curr Biol ; 13(2): R71-3, 2003 Jan 21.
Article in English | MEDLINE | ID: mdl-12546810

ABSTRACT

One critical step of apoptosis is the release of mitochondrial proteins through the outer mitochondrial membrane. Recent work shows that two pro-apoptotic Bcl-2 family proteins, Bax and Bid, as well as the mitochondrion-specific lipid cardiolipin may cooperate in chemically defined liposomes to generate a protein-permeable conduit, relaunching the debate on the identity of the pore responsible for mitochondrial membrane permeabilization during apoptosis.


Subject(s)
Apoptosis/physiology , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2 , Animals , BH3 Interacting Domain Death Agonist Protein , Cardiolipins/metabolism , Carrier Proteins/metabolism , Caspases/metabolism , In Vitro Techniques , Intracellular Membranes/metabolism , Matrix Metalloproteinases/metabolism , Models, Biological , Permeability , Proto-Oncogene Proteins/metabolism , bcl-2-Associated X Protein
20.
J Immunol Methods ; 265(1-2): 39-47, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12072177

ABSTRACT

Mitochondria undergo two major changes during early apoptosis. On the one hand, the outer mitochondrial membrane becomes permeable to proteins, resulting in the release of soluble intermembrane proteins (SIMPs) from the mitochondrion. On the other hand, the inner mitochondrial membrane transmembrane potential (DeltaPsi(m)) is reduced. These changes occur in most, if not all, models of cell death and can be taken advantage of to detect apoptosis at an early stage. Here, we delineate methods for the detection of alterations in the DeltaPsi(m), based on the incubation of cells with cationic lipophilic fluorochromes, the uptake of which is driven by the DeltaPsi(m). Certain DeltaPsi(m)-sensitive dyes can be combined with other fluorochromes to detect simultaneously cellular viability, plasma membrane exposure of phosphatidylserine residues, or the mitochondrial production of reactive oxygen species (ROS). In addition, we describe an immunofluorescence method for the detection of two functionally important proteins translocating from mitochondria, namely, the caspase co-activator cytochrome c and the caspase-independent death effector apoptosis inducing factor (AIF).


Subject(s)
Mitochondria/physiology , Animals , Apoptosis/physiology , Biological Transport , Humans , Membrane Potentials , Microscopy, Fluorescence , Reactive Oxygen Species
SELECTION OF CITATIONS
SEARCH DETAIL
...