Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
Add more filters










Publication year range
1.
J Cell Physiol ; 210(1): 177-82, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16998804

ABSTRACT

Previous studies from our laboratory indicate that cytosolic phospholipase A(2) (cPLA(2))-released arachidonic acid promotes monocyte/macrophage survival in the presence of peroxynitrite. In particular, the lipid messenger is metabolised by 5-lipoxygenase (5-LO) to 5-hydroxyeicosatetraenoic acid and causes the mitochondrial translocation of protein kinase Calpha (PKCalpha), an event associated with the cytosolic accumulation of Bad and Bax. Here we show that phosphorylation reactions driven by extracellular regulated kinase 1/2 (ERK1/2) critically regulate the activation/nuclear translocation of 5-LO. Inhibition of ERK1/2 was invariably associated with the cytosolic localisation of PKCalpha, the mitochondrial accumulation of Bad and Bax and with a rapid mitochondrial permeability transition-dependent necrosis. All these events were prevented by nanomolar concentrations of 5-hydroxyeicosatetraenoic acid. Hence, in addition to the previously characterised effects on cPLA(2), ERK1/2 critically regulates 5-LO activity in the absence of additional downstream targets in the survival signalling preventing peroxynitrite toxicity.


Subject(s)
Mitochondria/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/drug effects , Peroxynitrous Acid/pharmacology , Arachidonate 5-Lipoxygenase/metabolism , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Cell Survival/drug effects , Cyclosporine/pharmacology , Cytochromes c/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Hydroxyeicosatetraenoic Acids/metabolism , Hydroxyeicosatetraenoic Acids/pharmacology , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Monocytes/metabolism , Phospholipases A/antagonists & inhibitors , Phospholipases A/metabolism , Phosphorylation , Protein Kinase C-alpha/metabolism , Protein Transport/drug effects , U937 Cells , bcl-2-Associated X Protein/metabolism , bcl-Associated Death Protein/metabolism
2.
Cell Death Differ ; 11(9): 974-84, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15153939

ABSTRACT

We have studied the relationships existing between delayed formation of H2O2 and activation of cytosolic phospholipase A2 (cPLA2), events respectively promoting toxicity or survival in U937 cells exposed to peroxynitrite. The outcome of an array of different approaches using phospholipase A2 inhibitors, or cPLA2 antisense oligonucleotides, as well as specific respiratory chain inhibitors and respiration-deficient cells led to the demonstration that H2O2 does not mediate toxicity by producing direct molecular damage. Rather, the effects of H2O2 were found to be upstream to the arachidonic acid (AA)-mediated cytoprotective signalling and in fact causally linked to inhibition of cPLA2. Thus, it appears that U937 cells exposed to nontoxic concentrations of peroxynitrite are nevertheless committed to death, which however is normally prevented by the activation of parallel pathways resulting in cPLA2-dependent release of AA. A rapid necrotic response, however, takes place when high concentrations of peroxynitrite promote formation of H2O2 at levels impairing the cPLA2 cytoprotective signalling.


Subject(s)
Hydrogen Peroxide/pharmacology , Mitochondria , Peroxynitrous Acid/pharmacology , Phospholipases A/metabolism , Antimycin A/pharmacology , Arachidonic Acid/pharmacology , Blotting, Western , Catalase/metabolism , Cell Death , Cytosol/enzymology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Peroxide/metabolism , Immunohistochemistry , Microscopy, Confocal , Necrosis , Oligonucleotides , Oligonucleotides, Antisense/pharmacology , Oxygen Consumption , Phospholipases A2 , Rotenone/pharmacology , Signal Transduction , Temperature , Transfection , U937 Cells
3.
Cell Death Differ ; 9(12): 1368-76, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12478474

ABSTRACT

Peroxynitrite stimulates in U937 cells release of arachidonic acid (AA) sensitive to various phospholipase A(2) (PLA(2)) inhibitors, including arachidonyl trifluoromethyl ketone (AACOCF(3)), which specifically inhibits cytosolic PLA(2) (cPLA(2)). This response linearly increases using non toxic concentrations of the oxidant, and reaches a plateau at levels at which toxicity becomes apparent. Three separate lines of evidence are consistent with the notion that AA generated by cPLA(2) promotes survival in cells exposed to peroxynitrite. Firstly, toxicity was suppressed by nanomolar levels of exogenous AA, or by AA generated by the direct PLA(2) activator melittin. Secondly AACOCF(3), or other PLA(2) inhibitors, promoted cell death after exposure to otherwise non toxic concentrations of peroxynitrite; exogenous AA abolished the enhancing effects mediated by the PLA(2) inhibitors. Finally, U937 cells transfected with cPLA(2) antisense oligonucleotides were killed by concentrations of peroxynitrite that were non-toxic for cells transfected with nonsense oligonucleotides. This lethal response was insensitive to AACOCF(3) and prevented by exogenous AA.


Subject(s)
Arachidonic Acid/biosynthesis , Cell Death/physiology , Cell Survival/physiology , Cytosol/enzymology , Eukaryotic Cells/enzymology , Peroxynitrous Acid/metabolism , Phospholipases A/metabolism , Arachidonic Acids/pharmacology , Cell Death/drug effects , Cell Survival/drug effects , Cytosol/drug effects , Enzyme Inhibitors/pharmacology , Eukaryotic Cells/drug effects , Humans , Oligonucleotides, Antisense/pharmacology , Peroxynitrous Acid/pharmacology , Phospholipases A/drug effects , Phospholipases A/genetics , Phospholipases A2 , Tumor Cells, Cultured
4.
Cell Death Differ ; 9(11): 1248-55, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12404124

ABSTRACT

Activation of acid and neutral sphingomyelinases, and the ensuing generation of ceramide, contributes to the biological effects of tumour necrosis factor-alpha (TNF-alpha), one of which is apoptosis. While the mechanisms of activation of sphingomyelinases by the cytokine are being unravelled, less is known about regulation of their activity. Nitric oxide has previously been shown to exert a cyclic GMP-dependent inhibition of early apoptotic events triggered by TNF-alpha in the U937 monocytic cell line. We therefore investigated whether inhibition of sphingomyelinases by nitric oxide plays a role in regulating such early events. We found that activation of both acid and neutral sphingomyelinases, triggered in the first minutes after U937 cell stimulation with TNF-alpha, is regulated in an inhibitory fashion by nitric oxide, working through generation of cyclic GMP and activation of protein kinase G. Using a range of inhibitors selective for either sphingomyelinase we found that the acid sphingomyelinase contributes to activation of the initiator caspase-8 and early DNA fragmentation and that inhibition of the acid enzyme by nitric oxide accounts for cyclic GMP-dependent early protection from apoptosis. We also found that the protective effect by both cGMP and acid sphingomyelinase inhibitors progressively disappeared at later stages of the apoptotic process. Inhibition of sphingomyelinases represents a novel action of nitric oxide, which might be of physiological relevance in regulating initial phases of apoptosis as well as other biological actions of ceramide.


Subject(s)
Apoptosis/physiology , Cyclic GMP/metabolism , Nitric Oxide/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Apoptosis/drug effects , Cycloheximide/pharmacology , Humans , Protein Synthesis Inhibitors/pharmacology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
5.
Exp Cell Res ; 270(1): 56-65, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11597127

ABSTRACT

Treatment of U937 cells with nontoxic concentrations of TNFalpha increased the DNA strand scission induced by a short-chain lipid hydroperoxide analogue, tert-butylhydroperoxide. The following lines of evidence suggest that the enhancing effects of TNFalpha are mediated by inhibition of complex III and by the ensuing formation of superoxides and hydrogen peroxide: (a) the effects of TNFalpha were mimicked by the complex III inhibitor antimycin A; (b) the effects of TNFalpha, or antimycin A, were abolished by the complex I inhibitor rotenone, or by myxothiazol, an agent which inhibits the electron flow from the reduced coenzyme Q to cytochrome c(1) and therefore prevents ubisemiquinone formation; (c) the effects of TNFalpha, or antimycin A, were not observed in respiration-deficient cells; and (d) the effects of TNFalpha, or antimycin A, were sensitive to catalase. The TNFalpha-dependent inhibition of complex III appears to be mediated by ceramide. Three lines of evidence support this inference: (a) a synthetic cell-permeable ceramide analogue reproduced all the effects of TNFalpha, (b) TNFalpha promoted the formation of ceramide via a mechanism sensitive to inhibition of sphingomyelinases by tricyclodecan-9-yl-xanthogenate and imipramine, and (c) the TNFalpha-mediated enhancement of the tert-butylhydroperoxide-induced DNA-damaging response was prevented under conditions in which ceramide formation was inhibited.


Subject(s)
Ceramides/metabolism , DNA Damage , DNA, Single-Stranded/drug effects , Electron Transport Complex III/antagonists & inhibitors , Hydrogen Peroxide/metabolism , Sphingosine/analogs & derivatives , Superoxides/metabolism , Tumor Necrosis Factor-alpha/pharmacology , tert-Butylhydroperoxide/pharmacology , Antimycin A/pharmacology , Calcium/metabolism , Electron Transport , Enzyme Inhibitors/pharmacology , Humans , Mitochondria/metabolism , Sphingosine/pharmacology , Tumor Necrosis Factor-alpha/metabolism , U937 Cells , tert-Butylhydroperoxide/metabolism
6.
Eur J Biochem ; 268(20): 5223-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11606183

ABSTRACT

The short-chain lipid hydroperoxide analogue tert-butylhydroperoxide induces peroxynitrite-dependent and -independent DNA single strand breakage in PC12 cells. U937 cells that do not express constitutive nitric oxide synthase respond to tert-butylhydroperoxide treatment with peroxynitrite-independent DNA cleavage. Under experimental conditions leading to equivalent strand break frequencies, the analysis of poly(ADP-ribose) polymerase activity showed an increase in PC12 cells but not in U937 cells. The enhanced poly(ADP-ribose) polymerase activity observed in PC12 cells was paralleled by a significant decline in NAD+ content and both events were prevented by treatments suppressing formation of peroxynitrite. Although DNA breaks were rejoined at similar rates in the two cell lines, an inhibitor of poly(ADP-ribose) polymerase delayed DNA repair in PC12 cells but had hardly any effect in U937 cells. The results obtained using the latter cell type were confirmed with an additional cell line (Chinese hamster ovary cells) that does not express nitric oxide synthase. Collectively, our data suggest that tert-butylhydroperoxide-induced peroxynitrite-independent DNA strand scission is far less effective than the DNA cleavage generated by endogenous peroxynitrite in stimulating the activity of poly(ADP-ribose) polymerase.


Subject(s)
DNA Damage/drug effects , DNA, Single-Stranded/metabolism , Peroxynitrous Acid/metabolism , Poly(ADP-ribose) Polymerases/metabolism , tert-Butylhydroperoxide/pharmacology , Animals , Benzamides/pharmacology , CHO Cells , Cattle , Cricetinae , DNA Repair/drug effects , Enzyme Activation/drug effects , Humans , NAD/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , PC12 Cells , Rats , Time Factors , U937 Cells
7.
J Neurosci Res ; 65(5): 387-95, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11536321

ABSTRACT

A short-term exposure of PC12 cells to tert-butylhydroperoxide, followed by recovery in fresh culture medium, causes cell death and the extent of this response progressively increases during the 120 min of post-treatment incubation. Morphological and biochemical analyses of these cells revealed that the mode of cell death was necrosis. Cell killing induced by the hydroperoxide seems to be in part mediated by peroxynitrite because the lethal response was markedly and similarly reduced by the nitric oxide synthase inhibitor N omega-nitro-L-arginine methylester and by scavengers of nitric oxide or peroxynitrite. This peroxynitrite-dependent mechanism of cytotoxicity was blunted by antioxidants and inhibitors of mitochondrial permeability transition and the onset of cell death was preceded by mitochondrial depolarization and loss of cellular ATP. We conclude that tert-butylhydroperoxide promotes peroxynitrite-dependent PC12 cell necrosis causally linked to peroxidation of membrane lipids and mitochondrial permeability transition.


Subject(s)
Cell Death/drug effects , Cell Membrane Permeability/drug effects , Mitochondria/drug effects , Nitrates/pharmacology , PC12 Cells/drug effects , tert-Butylhydroperoxide/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cell Death/physiology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane Permeability/physiology , Dose-Response Relationship, Drug , Energy Metabolism/drug effects , Energy Metabolism/physiology , Enzyme Inhibitors/pharmacology , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Lipid Peroxides/metabolism , Membrane Lipids/metabolism , Mitochondria/metabolism , Necrosis , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurotoxins/metabolism , Neurotoxins/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , PC12 Cells/metabolism , PC12 Cells/pathology , Rats
8.
Free Radic Res ; 34(5): 513-27, 2001 May.
Article in English | MEDLINE | ID: mdl-11378534

ABSTRACT

A short term exposure to peroxynitrite promotes a time- and concentration-dependent lethal response in U937 cells. The mode of cell death was necrosis and rapid (within minutes) cell lysis was found to occur via a mechanism involving mitochondrial permeability transition. Apoptosis was not detected in cells exposed to low levels of peroxynitrite, or in cells which survived a treatment with toxic amounts of peroxynitrite, neither after the 60 min exposure nor following increasing time intervals of growth in fresh culture medium. Rather, cells treated with peroxynitrite concentrations which were not immediately lethal, as well as the survivors of treatments with toxic levels of peroxynitrite, proliferated with kinetics superimposable on those observed in untreated cells.


Subject(s)
Mitochondria/metabolism , Nitrates/pharmacology , Permeability , Antioxidants/pharmacology , Cell Division/drug effects , Cell Survival/drug effects , Chromans/pharmacology , Cyclosporine/pharmacology , Dose-Response Relationship, Drug , Humans , Intracellular Membranes/drug effects , Kinetics , Methionine/pharmacology , Mitochondria/drug effects , Necrosis , U937 Cells
9.
Biochem J ; 356(Pt 2): 509-13, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11368779

ABSTRACT

A well-established protocol to increase the intracellular content of ascorbic acid was used to investigate the effects of the vitamin on DNA single-strand breakage and toxicity mediated by authentic peroxynitrite (ONOO(-)) in U937 cells. This protocol involved exposure for 60 min to 100 microM dehydroascorbic acid, which was taken up by the cells and converted into ascorbic acid via a GSH-independent mechanism. At the time of exposure to ONOO(-), which was performed in fresh saline immediately after loading with dehydroascorbic acid, the vitamin present in the cells was all in its reduced form. It was found that, in cells that are otherwise ascorbate-deficient, an increase in their ascorbic acid content does not prevent, but rather enhances, the DNA-damaging and lethal responses mediated by exogenous ONOO(-). These results therefore suggest that acute supplementation of ascorbic acid can be detrimental for individuals with pathologies associated with a decrease in ascorbic acid and in which ONOO(-) is known to promote deleterious effects.


Subject(s)
Ascorbic Acid/metabolism , DNA Damage , Nitrates/toxicity , Ascorbic Acid/toxicity , Dehydroascorbic Acid/metabolism , Dehydroascorbic Acid/pharmacology , Ferricyanides/metabolism , Free Radical Scavengers/pharmacology , Humans , Iron Chelating Agents/pharmacology , Oxidation-Reduction , U937 Cells
10.
Free Radic Biol Med ; 29(8): 783-9, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11053780

ABSTRACT

A short-term exposure of PC12 cells to tert-butylhydroperoxide promotes a rapid oxidation of dihydrorhodamine sensitive to nitric oxide synthase inhibitors and peroxynitrite scavengers. This response was not directly caused by peroxynitrite, but rather appeared to be mediated by peroxynitrite-dependent activation of phospholipase A(2). The following lines of evidence support this inference: (i) the peroxynitrite-dependent dihydrorhodamine fluorescence response was blunted by low concentrations of two structurally unrelated phospholipase A(2) inhibitors; (ii) under similar conditions, the phospholipase A(2) inhibitors prevented release of arachidonic acid; (iii) low levels of arachidonic acid restored the dihydrorhodamine fluorescence response in nitric oxide synthase- as well as phospholipase A(2)-inhibited cells; (iv) the dihydrorhodamine fluorescence response induced by authentic peroxynitrite was also blunted by phospholipase A(2) inhibitors and restored upon addition of reagent arachidonic acid. We conclude that endogenous, or exogenous, peroxynitrite does not directly oxidize dihydrorhodamine in intact cells. Rather, peroxynitrite appears to act as a signalling molecule promoting release of arachidonic acid, which in turn leads to formation of species causing the dihydrorhodamine fluorescence response.


Subject(s)
Nitrates/metabolism , Nitrates/toxicity , Phospholipases A/metabolism , Animals , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Fluorescent Dyes , Microscopy, Confocal , Oxidants/metabolism , Oxidants/toxicity , PC12 Cells , Phospholipases A/antagonists & inhibitors , Rats , Reactive Oxygen Species/metabolism , Rhodamines , tert-Butylhydroperoxide/toxicity
11.
Free Radic Biol Med ; 28(11): 1619-27, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10938458

ABSTRACT

Both the phospholipase A(2) activator melittin and reagent arachidonic acid (AA) are poor inducers of DNA single strand breaks in U937 cells. These responses, however, were dramatically increased by the calcium-mobilizing agent caffeine (Cf) or by the respiratory substrate pyruvate via a mechanism that involved enforced mitochondrial Ca(2+) accumulation and that was sensitive to lipoxygenase inhibitors. In permeabilized cells, the DNA damage generated by AA in combination with either Cf, L-malate or CaCl(2) was blunted by catalase. AA generated DNA strand scission also in HeLa cells supplemented with pyruvate via a mechanism identical to that observed in U937 cells. This response was associated with an enforced formation of free radical species. These results demonstrate that mitochondria play a pivotal role in the DNA-damaging response evoked by AA and provide the bases for a calcium-dependent mechanism whereby the AA produced during inflammatory processes may affect various pathologic conditions, including carcinogenesis.


Subject(s)
Arachidonic Acid/pharmacology , Calcium/metabolism , DNA Damage , Mitochondria/drug effects , Mitochondria/metabolism , Caffeine/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Free Radicals/metabolism , HeLa Cells , Humans , Lipoxygenase Inhibitors/pharmacology , Malates/pharmacology , Pyruvic Acid/pharmacology , U937 Cells
12.
Proc Natl Acad Sci U S A ; 97(10): 5480-5, 2000 May 09.
Article in English | MEDLINE | ID: mdl-10792026

ABSTRACT

Apoptosis triggered by death receptors proceeds after defined signal-transduction pathways. Whether signaling at the receptor level is regulated by intracellular messengers is still unknown. We have investigated the role of two messengers, ceramide and nitric oxide (NO), on the apoptotic pathway activated in human monocytic U937 cells by tumor necrosis factor-alpha (TNF-alpha) working at its p55 receptor. Two transduction events, the receptor recruitment of the adapter protein, TRADD, and the activation of the initiator caspase, caspase 8, were investigated. When administered alone, neither of the messengers had any effect on these events. In combination with TNF-alpha, however, ceramide potentiated, whereas NO inhibited, TNF-alpha-induced TRADD recruitment and caspase 8 activity. The effect of NO, which was cGMP-dependent, was due to inhibition of the TNF-alpha-induced generation of ceramide. Our results identify a mechanism of regulation of a signal-transduction pathway activated by death receptors.


Subject(s)
Apoptosis/physiology , Ceramides/metabolism , Nitric Oxide/physiology , Penicillamine/analogs & derivatives , Tumor Necrosis Factor-alpha/pharmacology , Antigens, CD/physiology , Apoptosis/drug effects , Caspase 8 , Caspase 9 , Caspases/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP/physiology , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Nitric Oxide Donors/pharmacology , Oxadiazoles/pharmacology , Penicillamine/pharmacology , Proteins/metabolism , Quinoxalines/pharmacology , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type I , S-Nitroso-N-Acetylpenicillamine , Second Messenger Systems , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , TNF Receptor-Associated Factor 1 , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacokinetics , U937 Cells
13.
Br J Pharmacol ; 129(8): 1539-41, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10780956

ABSTRACT

A short term exposure of PC12 cells to a concentration of tert-butylhydroperoxide (tB-OOH) causing peroxynitrite-dependent DNA damage and cytotoxiticity promoted a release of arachidonic acid (AA) that was sensitive to phospholipase A(2) (PLA(2)) inhibitors and insensitive to phospholipase C or diacylglycerol lipase inhibitors. The extent of AA release was also mitigated by nitric oxide synthase (NOS) inhibitors and peroxynitrite scavengers. Low levels (10 microM) of authentic peroxynitrite restored the release of AA mediated by tB-OOH in NOS-inhibited cells whereas concentrations of peroxynitrite of 20 microM, or higher, effectively stimulated a PLA(2) inhibitor-sensitive release of AA also in the absence of additional treatments. These results are consistent with the possibility that endogenous as well as exogenous peroxynitrite promotes activation of PLA(2).


Subject(s)
Arachidonic Acid/metabolism , Nitrates/pharmacology , Animals , Oxidants/pharmacology , PC12 Cells , Phospholipases A/metabolism , Rats
14.
Eur J Neurosci ; 12(1): 145-54, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10651869

ABSTRACT

A short-term exposure to tert-butylhydroperoxide (tB-OOH) promoted a concentration-dependent formation of DNA single-strand breaks in PC12 cells. These events were paralleled by an increase in the cytosolic concentration of Ca2+ that was in part cleared by the mitochondria. Unlike the extent of Ca2+ mobilization and/or mitochondrial Ca2+ clearance, the DNA strand scission evoked by the hydroperoxide was markedly reduced by the nitric oxide (NO) scavenger 2-phenyl-4,4,5,5-tetramethylimidazolin-1-oxyl-3-oxide (PTIO) or by the NO synthase inhibitor N-nitro-L-arginine methylester (L-NAME). Inhibitors of electron transport (rotenone and myxothiazol), ruthenium red (RR, a polycation which inhibits the calcium uniporter of mitochondria), or peroxynitrite scavengers (Trolox and L-methionine) were as effective as PTIO or L-NAME in inhibiting the DNA-damaging response mediated by tB-OOH. Rotenone, RR or peroxynitrite scavengers did not further reduce the residual DNA cleavage observed following treatment with tB-OOH in L-NAME-supplemented cells. Exogenous NO also increased the DNA damage caused by tB-OOH in L-NAME-supplemented cells and this response was blunted by RR or by inhibitors of electron transport but was insensitive to peroxynitrite scavengers. We conclude that both endogenous and exogenous NO enhance the DNA cleavage generated by tB-OOH in PC12 cells. However, only endogenous NO set the bases for an involvement of peroxynitrite in this DNA-damaging response.


Subject(s)
Cyclic N-Oxides/pharmacology , DNA Damage/physiology , Imidazoles/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitrates/physiology , Nitric Oxide/physiology , tert-Butylhydroperoxide/pharmacology , Animals , Calcium/metabolism , Chromans/pharmacology , DNA Damage/drug effects , DNA, Single-Stranded/drug effects , Electron Transport/drug effects , Free Radical Scavengers/pharmacology , Methacrylates , Methionine/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Nitric Oxide Donors/pharmacology , Oxidants , PC12 Cells , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Rats , Rotenone/pharmacology , Ruthenium Red/pharmacology , S-Nitroso-N-Acetylpenicillamine , Thiazoles/pharmacology
15.
IUBMB Life ; 50(3): 195-201, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11142347

ABSTRACT

A large body of experimental evidence suggests that DNA damage and cytotoxicity mediated by peroxynitrite are linked by a causal relationship and important events in various pathological conditions. In the present study, we investigated the mechanism whereby peroxynitrite causes DNA single strand breakage in intact cells and found that the respiratory chain plays a pivotal role in this response. In particular, peroxynitrite mediates inhibition of complex III and, under these conditions, electrons are directly transferred from ubisemiquinone to molecular oxygen. Hydrogen peroxide produced by the dismutation of superoxides is the species mediating the peroxynitrite-dependent DNA cleavage.


Subject(s)
DNA Damage , Electron Transport/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Nitrates/pharmacology , Humans , Hydrogen Peroxide/metabolism , Kinetics , Methacrylates , Oxygen Consumption/drug effects , Potassium Cyanide/pharmacology , Rotenone/pharmacology , Thiazoles/pharmacology , U937 Cells
17.
Free Radic Res ; 33(5): 477-87, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11200081

ABSTRACT

Treatment of U937 cells with a sublethal concentration of tert-butylhydroperoxide generates DNA single strand breakage in U937 cells and this response is increased by caffeine, ATP, pyruvate or antimycin A. As we previously reported (Guidarelli, Clementi, Brambilla and Cantoni, (1997) Biochem. J. 328, 801-806), the enhancing effects of antimycin A are mediated by inhibition of complex III and the ensuing formation of superoxides and hydrogen peroxide in a reaction in which ubisemiquinone serves as an electron donor. Active electron transport was required in pyruvate-supplemented cells since the increased genotoxic response occurred as a consequence of enforced mitochondrial Ca2+ accumulation, a process driven by the increased electrochemical gradient. The enhancing effects of caffeine or ATP were also the consequence of mitochondrial Ca2+ accumulation but these responses were independent on electron transport. The increased formation of DNA lesions resulting from exposure to tert-butylhydroperoxide associated with the Ca2+-mobilizing agents or the respiratory substrate was mediated by arachidonic acid generated by Ca2+-dependent activation of phospholipase A2. Melittin, a potent phospholipase A2 activator, and reagent arachidonic acid mimicked the effects of caffeine, ATP or pyruvate on the tert-butylhydroperoxide-induced DNA single strand breakage.


Subject(s)
Arachidonic Acid/metabolism , Calcium/metabolism , DNA Damage , Mitochondria/metabolism , tert-Butylhydroperoxide/pharmacology , 5,8,11,14-Eicosatetraynoic Acid/pharmacology , Acetophenones/pharmacology , Antimycin A/pharmacology , Electron Transport , Enzyme Inhibitors/pharmacology , Humans , Melitten/pharmacology , Phospholipases A/antagonists & inhibitors , Phospholipases A2 , Quinacrine/pharmacology , U937 Cells
18.
Biochem Pharmacol ; 58(11): 1743-50, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10571248

ABSTRACT

A 3-hr exposure of U937 cells to hydrogen peroxide (H2O2) followed by a 6-hr posttreatment incubation in fresh culture medium promotes apoptosis or necrosis, depending on the oxidant concentration. Addition of 3-aminobenzamide (3AB) during the recovery phase prevented necrosis and caused apoptosis. 3AB did not, however, affect the apoptotic response of cells treated with apogenic concentrations of H2O2. Cells exposed for 3 hr to 1.5 mM H2O2, while showing some signs of suffering, maintained a normal nuclear organization and good organelle morphology. At the biochemical level, the oxidant promoted the formation of Mb-sized DNA fragments and rapidly depleted both the adenine nucleotide and non-protein sulphydryl pools, which did not recover during posttreatment incubation in the absence or presence of 3AB. These results allow a novel interpretation of the concentration-dependent switch from apoptosis to necrosis. We propose that H2O2 activates the apoptotic response at the early times of peroxide exposure and that this process can be completed, or inhibited, during the posttreatment incubation phase. Inhibition of apoptosis leads to necrosis and can be prevented by 3AB via a mechanism independent of inhibition of poly(ADP-ribose)polymerase. As a corollary, the necrotic response promoted by high concentrations of H2O2 in U937 cells appears to be the result of specific inhibition of the late steps of apoptosis.


Subject(s)
Apoptosis , Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Hydrogen Peroxide/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Adenine Nucleotides/metabolism , DNA/metabolism , DNA Fragmentation/drug effects , Drug Interactions , Humans , Necrosis , Oxidants/pharmacology , U937 Cells
19.
Free Radic Res ; 31(2): 93-101, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10490238

ABSTRACT

Exposure of U937 cells to the antioxidant 2,6-di-tert-butyl-4-methylphenol (BHT), unlike exposure to other antioxidants such as N,N'-diphenyl-1,4-phenylenediamine, Trolox or alpha-tocopherol, promotes a time- and concentration-dependent induction of apoptosis. This response was prevented by the iron chelator o-phenanthroline and by the thiol reagent N-acetylcysteine but was increased remarkably in cells pre-exposed to the catalase inhibitor 3-amino-1,2,4-triazole or to L-buthionine-[S,R]-sulfoximine, a specific inhibitor of glutathione synthesis. Furthermore, the BHT-induced apoptotic response was markedly enhanced by cytochrome P450 inhibitors. Taken together, the experimental results presented in this study indicate that BHT efficiently induces apoptosis in U937 cells and that this response is not caused by products of cytochrome P450 metabolism. Instead, apoptosis appeared to be causally linked to an altered cellular redox state in which hydrogen peroxide plays a pivotal role.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Butylated Hydroxytoluene/pharmacology , Hydrogen Peroxide/metabolism , Oxidation-Reduction , Acetylcysteine/pharmacology , Buthionine Sulfoximine/pharmacology , Catalase/metabolism , Cytochrome P-450 Enzyme Inhibitors , DNA Fragmentation , Drug Combinations , Drugs, Chinese Herbal/pharmacology , Electrophoresis, Agar Gel , Enzyme Inhibitors/pharmacology , Free Radical Scavengers , Glycyrrhiza , Humans , Iron Chelating Agents/pharmacology , Metyrapone/pharmacology , Paeonia , Phenanthrolines/pharmacology , U937 Cells
20.
FEBS Lett ; 457(1): 139-43, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10486581

ABSTRACT

Exposure of U937 cells to tert-butylhydroperoxide (tB-OOH) led to cyclosporin A-sensitive mitochondrial membrane permeability transition and necrosis. Pyruvate and rotenone, which increase mitochondrial NADH via different mechanisms, prevented these responses and the cells which received these treatments proliferated with kinetics similar to those observed in untreated cells. In contrast with these results, cells rescued by cyclosporin A were unable to proliferate. Thus, mitochondrial NADH plays a pivotal role in preventing upstream events which result in the onset of mitochondrial membrane permeability transition and death in cells exposed to tB-OOH. These events appear to be critical for recovery of the ability of the cells to proliferate.


Subject(s)
Cell Division/drug effects , Pyruvic Acid/pharmacology , Rotenone/pharmacology , Uncoupling Agents/pharmacology , tert-Butylhydroperoxide/pharmacology , Apoptosis , Cell Count , Cell Survival , Cyclosporine/pharmacology , DNA Fragmentation , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Necrosis , Time Factors , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...