Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 22(11): 1877-89, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25909891

ABSTRACT

Neurotransmission unavoidably increases mitochondrial reactive oxygen species. However, the intrinsic antioxidant defense of neurons is weak and hence the mechanism whereby these cells are physiologically protected against oxidative damage is unknown. Here we found that the antioxidant defense of neurons is repressed owing to the continuous protein destabilization of the master antioxidant transcriptional activator, nuclear factor-erythroid 2-related factor-2 (Nrf2). By contrast, Nrf2 is highly stable in neighbor astrocytes explaining their robust antioxidant defense and resistance against oxidative stress. We also show that subtle and persistent stimulation of N-methyl-d-aspartate receptors (NMDAR) in astrocytes, through a mechanism not requiring extracellular Ca²âº influx, upregulates a signal transduction pathway involving phospholipase C-mediated endoplasmic reticulum release of Ca²âº and protein kinase Cδ activation. Active protein kinase Cδ promotes, by phosphorylation, the stabilization of p35, a cyclin-dependent kinase-5 (Cdk5) cofactor. Active p35/Cdk5 complex in the cytosol phosphorylates Nrf2 at Thr(395), Ser(433) and Thr(439) that is sufficient to promote Nrf2 translocation to the nucleus and induce the expression of antioxidant genes. Furthermore, this Cdk5-Nrf2 transduction pathway boosts glutathione metabolism in astrocytes efficiently protecting closely spaced neurons against oxidative damage. Thus, intercellular communication through NMDAR couples neurotransmission with neuronal survival.


Subject(s)
Astrocytes/metabolism , Cyclin-Dependent Kinase 5/metabolism , NF-E2-Related Factor 2/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Astrocytes/drug effects , Blotting, Western , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Cyclin-Dependent Kinase 5/genetics , Flow Cytometry , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Immunoprecipitation , Lipids/pharmacology , Membrane Potential, Mitochondrial/drug effects , NF-E2-Related Factor 2/genetics , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction
3.
Cell Death Differ ; 19(10): 1582-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22421967

ABSTRACT

6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3) is a master regulator of glycolysis by its ability to synthesize fructose-2,6-bisphosphate, a potent allosteric activator of 6-phosphofructo-1-kinase. Being a substrate of the E3 ubiquitin ligase anaphase-promoting complex-Cdh1 (APC(Cdh1)), PFKFB3 is targeted to proteasomal degradation in neurons. Here, we show that activation of N-methyl-D-aspartate subtype of glutamate receptors (NMDAR) stabilized PFKFB3 protein in cortical neurons. Expressed PFKFB3 was found to be mainly localized in the nucleus, where it is subjected to degradation; however, expression of PFKFB3 lacking the APC(Cdh1)-targeting KEN motif, or following NMDAR stimulation, promoted accumulation of PFKFB3 and its release from the nucleus to the cytosol through an excess Cdh1-inhibitable process. NMDAR-mediated increase in PFKFB3 yielded neurons having a higher glycolysis and lower pentose-phosphate pathway (PPP); this led to oxidative stress and apoptotic neuronal death that was counteracted by overexpressing glucose-6-phosphate dehydrogenase, the rate-limiting enzyme of the PPP. Furthermore, expression of the mutant form of PFKFB3 lacking the KEN motif was sufficient to trigger oxidative stress and apoptotic death of neurons. These results reveal that, by inhibition of APC(Cdh1), glutamate receptors activation stabilizes PFKFB3 thus switching neuronal metabolism leading to oxidative damage and neurodegeneration.


Subject(s)
Phosphofructokinase-2/metabolism , Amino Acid Motifs , Anaphase-Promoting Complex-Cyclosome , Animals , Apoptosis/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Glucosephosphate Dehydrogenase/genetics , Glucosephosphate Dehydrogenase/metabolism , Glutamic Acid/pharmacology , Glycolysis/drug effects , Mutagenesis, Site-Directed , N-Methylaspartate/pharmacology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Pentose Phosphate Pathway/drug effects , Phosphofructokinase-2/antagonists & inhibitors , Phosphofructokinase-2/chemistry , Phosphofructokinase-2/genetics , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism
4.
Oncogene ; 27(23): 3339-44, 2008 May 22.
Article in English | MEDLINE | ID: mdl-18212744

ABSTRACT

In neuroblastoma cells, retinoic acid induces cell cycle arrest and differentiation through degradation of the F-box protein, Skp2, and stabilization of cyclin-dependent kinase inhibitor, p27. However, the mechanism responsible for retinoic acid-mediated Skp2 destabilization is unknown. Since Skp2 is degraded by anaphase-promoting complex (APC)(Cdh1), here we studied whether retinoic acid promotes differentiation of human SH-SY5Y neuroblastoma cells by modulating Cdh1. We found that retinoic acid induced the nuclear accumulation of Cdh1 that paralleled Skp2 destabilization and p27 accumulation. The mRNA and protein abundance of Rae1-a nuclear export factor that limits APC(Cdh1) activity in mitosis-decreased upon retinoic acid-induced inhibition of neuroblastoma cell proliferation. Furthermore, either Rae1 overexpression or Cdh1 inhibition promoted Skp2 accumulation, p27 destabilization and prevented retinoic acid-induced cell cycle arrest and differentiation. Conversely, inhibition of Rae1 accelerated retinoic acid-induced differentiation. Thus, retinoic acid downregulates Rae1, hence facilitating APC(Cdh1)-mediated Skp2 degradation leading to the arrest of cell cycle progression and neuroblastoma differentiation.


Subject(s)
Cell Differentiation/drug effects , Neuroblastoma/pathology , Nuclear Matrix-Associated Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Tretinoin/pharmacology , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neuroblastoma/genetics , Nuclear Matrix-Associated Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Protein Processing, Post-Translational/drug effects , S-Phase Kinase-Associated Proteins/metabolism
5.
Biochem Soc Trans ; 35(Pt 5): 1224-7, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17956318

ABSTRACT

NO (nitric oxide) participates in a considerable number of physiological functions. At the biochemical level, most of its actions can be ascribed to its ability to bind, and activate, soluble guanylate cyclase. However, mounting evidence now strongly suggests that the NO-mediated inhibition of cytochrome c oxidase, the terminal complex of the mitochondrial respiratory chain, may be a further step of a cell signalling process involved in the regulation of important cellular functions. In most cells, including neurons and astrocytes, NO reversibly, and irreversibly, modulates O(2) consumption, a phenomenon through which NO signals certain pathways relevant for neuronal survival. Here, we propose that besides the control of mitochondrial bioenergetics, NO finely modulates the balance between glucose consumption through the glycolytic pathway and the pentose phosphate pathway in neurons. This may have implications for our understanding of the mechanisms of neurodegeneration due to oxidative and nitrosative stress.


Subject(s)
Neurons/physiology , Nitric Oxide/physiology , Oxidative Stress , Animals , Glycolysis , Mitochondria/physiology , Neurons/enzymology , Neurons/metabolism , Oxidation-Reduction
6.
Cell Death Differ ; 14(6): 1211-21, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17347665

ABSTRACT

In neurons, DNA is prone to free radical damage, although repair mechanisms preserve the genomic integrity. However, activation of the DNA repair system, poly(ADP-ribose) polymerase (PARP-1), is thought to cause neuronal death through NAD+ depletion and mitochondrial membrane potential (delta psi(m)) depolarization. Here, we show that abolishing PARP-1 activity in primary cortical neurons can either enhance or prevent apoptotic death, depending on the intensity of an oxidative stress. Only in severe oxidative stress does PARP-1 activation result in NAD+ and ATP depletion and neuronal death. To investigate the role of PARP-1 in an endogenous model of oxidative stress, we used an RNA interference (RNAi) strategy to specifically knock down glutamate-cysteine ligase (GCL), the rate-limiting enzyme of glutathione biosynthesis. GCL RNAi spontaneously elicited a mild type of oxidative stress that was enough to stimulate PARP-1 in a Ca2+-calmodulin kinase II-dependent manner. GCL RNAi-mediated PARP-1 activation facilitated DNA repair, although neurons underwent delta psi(m) loss followed by some apoptotic death. PARP-1 inhibition did not prevent delta psi(m) loss, but enhanced the vulnerability of neurons to apoptosis upon GCL silencing. Conversely, mild expression of PARP-1 partially prevented to GCL RNAi-dependent apoptosis. Thus, in the mild progressive damage likely occur in neurodegenerative diseases, PARP-1 activation plays a neuroprotective role that should be taken into account when considering therapeutic strategies.


Subject(s)
Apoptosis/physiology , Neurons/metabolism , Oxidative Stress/physiology , Poly(ADP-ribose) Polymerases/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , DNA Repair , Dose-Response Relationship, Drug , Flow Cytometry , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Humans , Hydrogen Peroxide/pharmacology , Immunohistochemistry , In Situ Nick-End Labeling , Mutagenesis, Site-Directed , NAD/metabolism , Neurons/cytology , Neurons/drug effects , Poly(ADP-ribose) Polymerases/genetics , RNA Interference , Rats , Rats, Inbred WF
7.
J Neurochem ; 86(1): 228-37, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12807442

ABSTRACT

Primary culture rat astrocytes exposed to the long acting nitric oxide donor (Z)-1-[2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate (DETA-NO) for 24 h approximately double their concentration of glutathione (GSH) and show no sign of cell death. In contrast, GSH was depleted by 48%, and significant loss of mitochondrial respiratory chain complex activity and cell death were observed in primary culture rat neurones subjected to DETA-NO for 18 h. Northern blot analysis suggested that mRNA amounts of both subunits of glutamate-cysteine ligase (GCL), the rate-limiting enzyme in GSH synthesis, were elevated in astrocytes following nitric oxide (NO) exposure. This correlated with an increase in astrocytic GCL activity. Neurones on the other hand did not exhibit increased GCL activity when exposed to NO. In addition, the rate of GSH efflux was doubled and gamma-glutamyltranspeptidase (gamma-GT) activity was increased by 42% in astrocytes treated with NO for 24 h. These results suggest that astrocytes, but not neurones, up-regulate GSH synthesis as a defence mechanism against excess NO. It is possible that the increased rate of GSH release and activity of gamma-GT in astrocytes may have important implications for neuroprotection in vivo by optimizing the supply of GSH precursors to neurones in close proximity.


Subject(s)
Astrocytes/metabolism , Glutathione/metabolism , Mitochondria/drug effects , Neurons/metabolism , Nitric Oxide/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cells, Cultured , Electron Transport/drug effects , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/etiology , Neurons/cytology , Neurons/drug effects , Neuroprotective Agents , Nitric Oxide/pharmacology , Nitric Oxide Donors/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Triazenes/pharmacology , gamma-Glutamyltransferase/drug effects , gamma-Glutamyltransferase/metabolism
8.
Proc Natl Acad Sci U S A ; 98(26): 15294-9, 2001 Dec 18.
Article in English | MEDLINE | ID: mdl-11742096

ABSTRACT

It was recently proposed that in Jurkat cells, after inhibition of respiration by NO, glycolytically generated ATP plays a critical role in preventing the collapse of mitochondrial membrane potential (Deltapsi(m)) and thus apoptotic cell death. We have investigated this observation further in primary cultures of rat cortical neurons and astrocytes-cell types that differ greatly in their glycolytic capacity. Continuous and significant ( approximately 85%) inhibition of respiration by NO (1.4 microM at 175 microM O(2)) generated by [(z)-1-[2-aminoethyl]-N-[2-ammonioethyl]amino]diazen-1-ium-1,2 diolate (DETA-NO) initially (10 min) depleted ATP concentrations by approximately 25% in both cell types and increased the rate of glycolysis in astrocytes but not in neurons. Activation of glycolysis in astrocytes, as judged by lactate production, prevented further ATP depletion, whereas in neurons, which do not invoke this mechanism, there was a progressive decrease in ATP concentrations over the next 60 min. During this time, there was a persistent mitochondrial hyperpolarization and absence of apoptotic cell death in astrocytes, whereas in the neurons there was a progressive fall in Deltapsi(m) and increased apoptosis. After glucose deprivation or treatment with inhibitors of the F(1)F(0)-ATPase and adenine nucleotide translocase, astrocytes responded to NO with a fall in Deltapsi(m) and apoptotic cell death similar to the response in neurons. Finally, although treatment of astrocytes with NO partially prevented staurosporin-induced collapse in Deltapsi(m) and cell death, NO and staurosporin synergized in decreasing Deltapsi(m) and inducing apoptosis in neurons. These results demonstrate that although inhibition of cellular respiration by NO leads to neurotoxicity, it may also result in initial neuroprotection, depending on the glycolytic capacity of the particular cell.


Subject(s)
Adenosine Triphosphate/metabolism , Astrocytes/drug effects , Neurons/drug effects , Nitric Oxide/pharmacology , Animals , Apoptosis/drug effects , Astrocytes/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Glycolysis , Membrane Potentials , Mitochondria/physiology , Neurons/physiology , Oxygen Consumption , Rats , Rats, Wistar , Staurosporine/pharmacology
9.
J Neurochem ; 79(1): 17-24, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11595753

ABSTRACT

The induction of nitric oxide (NO) synthase in astrocytes by endotoxin and/or cytokine treatment is associated with increased glucose consumption and glycolysis, but the mechanism whereby this phenomenon occurs remains obscure. In this work, we have addressed this issue and found that incubation of cultured rat astrocytes with lipopolysaccharide (LPS; 1 microg/mL) for 24 h increased the level of constitutively expressed GLUT1 glucose transporter mRNA, and triggered GLUT3 mRNA expression, which was absent in normal astrocytes. The occurrence of GLUT3 protein after LPS treatment was corroborated by western blotting and immunocytochemistry. A 4-h incubation of astrocytes in the absence of glucose, or under an oxygen-poor (3%) atmosphere also resulted in GLUT3 mRNA overexpression. Experiments performed with 2-deoxy-D-[U-14C]glucose (at 0.1 mM of D-glucose) confirmed that LPS (0.1-10 microg/mL) dose-dependently increased the rate of glucose uptake (by a factor of 1.6 at 1 microg/mL of LPS), which was paralleled with the increase in NO synthesis. Furthermore, blockade of NO synthase with 2-amino-5,6-dihydro-6-methyl-(4H)-1,3-thiazine (AMT; 50 microM) partially (by 45%) prevented the LPS-mediated increase in glucose uptake. Finally, incubation of astrocytes with the NO donor 1-[2-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate (DETA; 100 microM) increased by a factor of 1.4 the rate of glucose uptake. We conclude that the increase in GLUT3-driven glucose uptake in astrocytes would have a neuroprotective role under conditions in which NO formation is combined with hypoglycaemia, such as in brain ischemia.


Subject(s)
Astrocytes/metabolism , Gene Expression , Lipopolysaccharides/pharmacology , Monosaccharide Transport Proteins/genetics , Nerve Tissue Proteins , Nitric Oxide/physiology , Animals , Animals, Newborn , Blotting, Western , Cell Hypoxia , Cells, Cultured , Glucose/administration & dosage , Glucose/metabolism , Glucose Transporter Type 1 , Glucose Transporter Type 3 , Immunohistochemistry , Kinetics , NF-kappa B/metabolism , Nitric Oxide Donors/pharmacology , RNA, Messenger/analysis , Rats , Rats, Wistar
11.
J Neurochem ; 77(2): 676-90, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11299330

ABSTRACT

In order to investigate the relationship between nitric oxide-mediated regulation of mitochondrial function and excitotoxicity, the role of mitochondrial ATP synthesis and intracellular redox status on the mode of neuronal cell death was studied. Brief (5 min) glutamate (100 microM) receptor stimulation in primary cortical neurons collapsed the mitochondrial membrane potential (psi(m)) and transiently (30 min) inhibited mitochondrial ATP synthesis, causing early (1 h) necrosis or delayed (24 h) apoptosis. The transient inhibition of ATP synthesis was paralleled to a loss of NADH, which was fully recovered shortly after the insult. In contrast, NADPH and the GSH/GSSG ratio were maintained, but progressively decreased thereafter. Twenty-four hours after glutamate treatment, ATP was depleted, a phenomenon associated with a persistent inhibition of mitochondrial succinate-cytochrome c reductase activity and delayed necrosis. Blockade of either nitric oxide synthase (NOS) activity or the mitochondrial permeability transition (MPT) pore prevented psi(m) collapse, the transient inhibition of mitochondrial ATP synthesis, early necrosis and delayed apoptosis. However, blockade of NOS activity, but not the MPT pore, prevented the inhibition of succinate-cytochrome c reductase activity and delayed ATP depletion and necrosis. From these results, we suggest that glutamate receptor-mediated NOS activation would trigger MPT pore opening and transient inhibition of ATP synthesis leading to apoptosis in a neuronal subpopulation, whereas other groups of neurons would undergo oxidative stress and persistent inhibition of ATP synthesis leading to necrosis.


Subject(s)
Adenosine Triphosphate/physiology , Apoptosis/drug effects , Cerebral Cortex/cytology , Glutamic Acid/pharmacology , Neurons/drug effects , Neurotoxins/pharmacology , 2-Amino-5-phosphonovalerate/pharmacology , Adenosine Triphosphate/biosynthesis , Animals , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cyclosporine/pharmacology , Electron Transport/drug effects , Electron Transport Complex I , Energy Metabolism/drug effects , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , NAD/metabolism , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NG-Nitroarginine Methyl Ester/pharmacology , Necrosis , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Oligomycins/pharmacology , Permeability/drug effects , Rats , Rats, Wistar , Receptors, Glutamate/drug effects , Receptors, Glutamate/physiology , Rotenone/pharmacology , Single-Blind Method , Succinate Cytochrome c Oxidoreductase/antagonists & inhibitors , omega-N-Methylarginine/pharmacology
12.
J Neurochem ; 76(5): 1593-6, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238744

ABSTRACT

Glutathione deficiency is commonly associated with mitochondrial complex I dysfunction and loss of viability in neurones, but not in glia. In order to address the possible mechanism responsible for this cellular difference, the regulation of mitochondrial complex I expression by glutathione depletion was investigated in glial cells. Incubation of rat-cultured astrocytes and C6 glioma cells with the specific gamma-glutamylcysteine synthetase inhibitor L-buthionine-(S:,R:)-sulfoximine (L-BSO; 0.1-1 mM) decreased the total specific content of glutathione in a dose- and time-dependent fashion. Northern blot analyses revealed that glutathione deficiency caused by L-BSO (0.1 mM) was associated with a twofold enhancement in complex I regulatory subunit ND6 (mitochondrially encoded) mRNA expression after 24-72 h. This effect was accompanied by a twofold increase in complex-I activity at 72 h in L-BSO-treated cells, as compared with control cells, but complex II-III, complex IV and citrate synthase activities were unaltered. It is suggested that the oxidative stress caused by glutathione depletion in glial cells would up-regulate complex-I activity by enhancing the expression of the mitochondrially encoded regulatory subunit. These results could offer further insight into the different degree of cellular susceptibility observed in glial vs. neuronal cells against oxidative stress.


Subject(s)
Astrocytes/enzymology , Buthionine Sulfoximine/pharmacology , Gene Expression Regulation, Enzymologic , Glutathione/metabolism , NADH, NADPH Oxidoreductases/genetics , Neuroglia/enzymology , Animals , Animals, Newborn , Cells, Cultured , Citrate (si)-Synthase/genetics , Electron Transport Complex I , Electron Transport Complex II , Gene Expression Regulation, Enzymologic/drug effects , Glioma , Multienzyme Complexes/genetics , Oxidoreductases/genetics , Prosencephalon/cytology , Prosencephalon/enzymology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Succinate Dehydrogenase/genetics , Tumor Cells, Cultured
13.
J Neurochem ; 75(4): 1618-24, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10987843

ABSTRACT

The possible neuroprotective effect of D-glucose against glutamate-mediated neurotoxicity was studied in rat cortical neurons in primary culture. Brief (5-min) exposure of neurons to glutamate (100 microM) increased delayed (24-h) necrosis and apoptosis by 3- and 1.8-fold, respectively. Glutamate-mediated neurotoxicity was accompanied by a D-(-)-2-amino-5-phosphonopentanoate (100 microM) and N(omega)-nitro-L-arginine methyl ester (1 mM)-inhibitable, time-dependent ATP depletion (55% at 24 h), confirming the involvement of NMDA receptor stimulation followed by nitric oxide synthesis in this process. Furthermore, the presence of D-glucose (20 mM), but not its inactive enantiomer, L-glucose, fully prevented glutamate-mediated delayed ATP depletion, necrosis, and apoptosis. Succinate- cytochrome c reductase activity, but not the activities of NADH-coenzyme Q(1) reductase or cytochrome c oxidase, was inhibited by 32% by glutamate treatment, an effect that was abolished by incubation with D-glucose. Lactate accumulation in the culture medium was unmodified by any of these treatments, ruling out the possible involvement of the glycolysis pathway in either glutamate neurotoxicity or D-glucose neuroprotection. In contrast, D-glucose, but not L-glucose, abolished glutamate-mediated glutathione oxidation and NADPH depletion. Our results suggest that NADPH production from D-glucose accounts for glutathione regeneration and protection from mitochondrial dysfunction. This supports the notion that the activity of the pentose phosphate pathway may be an important factor in protecting neurons against glutamate neurotoxicity.


Subject(s)
Glucose/metabolism , Glutathione/metabolism , Mitochondria/metabolism , Neurons/metabolism , Receptors, Glutamate/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Death , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Energy Metabolism/drug effects , Glucose/pharmacology , Lactic Acid/metabolism , Mitochondria/drug effects , NAD/metabolism , Neurons/cytology , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide/biosynthesis , Oxidation-Reduction/drug effects , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism
14.
J Neurochem ; 73(4): 1446-52, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10501188

ABSTRACT

The biosynthesis of the physiological messenger nitric oxide (*NO) in neuronal cells is thought to depend on a glial-derived supply of the *NO synthase substrate arginine. To expand our knowledge of the mechanism responsible for this glial-neuronal interaction, we studied the possible roles of peroxynitrite anion (ONOO-), superoxide anion (O2*-), *NO, and H2O2 in L-[3H]arginine release in cultured rat astrocytes. After 5 min of incubation at 37 degrees C, initial concentrations of 0.05-2 mM ONOO- stimulated the release of arginine from astrocytes in a concentration-dependent way; this effect was maximum from 1 mM ONOO- and proved to be approximately 400% as compared with control cells. ONOO(-)-mediated arginine release was prevented by arginine transport inhibitors, such as L-lysine and N(G)-monomethyl-L-arginine, suggesting an involvement of the arginine transporter in the effect of ONOO-. In situ xanthine/xanthine oxidase-generated O2*- (20 nmol/min) stimulated arginine release to a similar extent to that found with 0.1 mM ONOO-, but this effect was not prevented by arginine transport inhibitors. *NO donors, such as sodium nitroprusside, S-nitroso-N-acetylpenicillamine, or 1-[2-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium+ ++-1,2-diolate, and H2O2 did not significantly modify arginine release. As limited arginine availability for neuronal *NO synthase activity may be neurotoxic due to ONOO- formation, our results suggest that ONOO(-)-mediated arginine release from astrocytes may contribute to replenishing neuronal arginine, hence avoiding further generation of ONOO- within these cells.


Subject(s)
Arginine/metabolism , Astrocytes/physiology , Nitrates/pharmacology , Oxidants/pharmacology , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/drug effects , Cells, Cultured , Citrulline/metabolism , Glutamic Acid/metabolism , Hydrogen Peroxide/pharmacology , Kinetics , Lysine/pharmacology , Nitric Oxide Donors/pharmacology , Rats , Rats, Wistar , Serine/metabolism
15.
FEBS Lett ; 452(3): 290-4, 1999 Jun 11.
Article in English | MEDLINE | ID: mdl-10386608

ABSTRACT

The possible role of nitric oxide (*NO) in brain mitochondrial maturation was studied. Within the first 5 min after birth, a sharp increase in ATP concentrations was observed, coinciding with an increase in mitochondrial complex II-III (succinate-cytochrome c reductase) activity, while complex I (NADH-CoQ1 reductase) and complex IV (cytochrome c oxidase) activities remained unchanged. Under the same circumstances, cGMP concentrations were increased by 5 min after birth, correlating significantly with ATP concentrations. Since ATP concentrations also correlated significantly with mitochondrial complex II-III activity, these three parameters may be associated. Inhibition of *NO synthase activity brought about by the administration of N(omega)-nitro-L-arginine monomethyl ester to mothers prevented the postnatal increase in cGMP and ATP levels and complex II-III activity. These results suggest that early postnatal mitochondrial maturation in the brain is a *NO-mediated process.


Subject(s)
Adenosine Triphosphate/metabolism , Aging/physiology , Brain/metabolism , Mitochondria/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/physiology , Animals , Animals, Newborn , Brain/drug effects , Brain/growth & development , Citrate (si)-Synthase/metabolism , Cyclic GMP/metabolism , Electron Transport Complex II , Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Mitochondria/drug effects , Multienzyme Complexes/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Nitric Oxide Synthase/metabolism , Oxidoreductases/metabolism , Rats , Rats, Wistar , Succinate Dehydrogenase/metabolism
16.
Biochim Biophys Acta ; 1411(2-3): 415-36, 1999 May 05.
Article in English | MEDLINE | ID: mdl-10320673

ABSTRACT

A large body of evidence has appeared over the last 6 years suggesting that nitric oxide biosynthesis is a key factor in the pathophysiological response of the brain to hypoxia-ischemia. Whilst studies on the influence of nitric oxide in this phenomenon initially offered conflicting conclusions, the use of better biochemical tools, such as selective inhibition of nitric oxide synthase (NOS) isoforms or transgenic animals, is progressively clarifying the precise role of nitric oxide in brain ischemia. Brain ischemia triggers a cascade of events, possibly mediated by excitatory amino acids, yielding the activation of the Ca2+-dependent NOS isoforms, i.e. neuronal NOS (nNOS) and endothelial NOS (eNOS). However, whereas the selective inhibition of nNOS is neuroprotective, selective inhibition of eNOS is neurotoxic. Furthermore, mainly in glial cells, delayed ischemia or reperfusion after an ischemic episode induces the expression of Ca2+-independent inducible NOS (iNOS), and its selective inhibition is neuroprotective. In conclusion, it appears that activation of nNOS or induction of iNOS mediates ischemic brain damage, possibly by mitochondrial dysfunction and energy depletion. However, there is a simultaneous compensatory response through eNOS activation within the endothelium of blood vessels, which mediates vasodilation and hence increases blood flow to the damaged brain area.


Subject(s)
Brain Ischemia/physiopathology , Brain/metabolism , Hypoxia, Brain/physiopathology , Nitric Oxide/metabolism , Animals , Brain/enzymology , Excitatory Amino Acids/metabolism , Humans , Nitrates/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type III , Perinatology , Vasodilation
17.
FEBS Lett ; 446(2-3): 261-3, 1999 Mar 12.
Article in English | MEDLINE | ID: mdl-10100854

ABSTRACT

The assumption that reversible inhibition of mitochondrial respiration by nitric oxide (NO.) represents inhibition of ATP synthesis is unproven. NO. could theoretically inhibit the oxygen consumption with continued ATP synthesis, by acting as an electron acceptor from cytochrome c or as a terminal electron acceptor in stead of oxygen. We report here that NO. does reversibly inhibit brain mitochondrial ATP synthesis with a time course similar to its inhibition of respiration. Whilst such inhibition was largely reversible, there appeared to be a small irreversible component which may theoretically be due to peroxynitrite formation, i.e. as a result of the reaction between NO. and superoxide, generated by the mitochondrial respiratory chain.


Subject(s)
Adenosine Triphosphate/biosynthesis , Nitric Oxide/metabolism , Adenosine Triphosphate/antagonists & inhibitors , Animals , Male , Mitochondria/drug effects , Mitochondria/metabolism , Nitric Oxide/pharmacology , Rats , Rats, Wistar
18.
Biochim Biophys Acta ; 1410(2): 215-28, 1999 Feb 09.
Article in English | MEDLINE | ID: mdl-10076028

ABSTRACT

Damage to the mitochondrial electron transport chain has been suggested to be an important factor in the pathogenesis of a range of neurological disorders, such as Parkinson's disease, Alzheimer's disease, multiple sclerosis, stroke and amyotrophic lateral sclerosis. There is also a growing body of evidence to implicate excessive or inappropriate generation of nitric oxide (NO) in these disorders. It is now well documented that NO and its toxic metabolite, peroxynitrite (ONOO-), can inhibit components of the mitochondrial respiratory chain leading, if damage is severe enough, to a cellular energy deficiency state. Within the brain, the susceptibility of different brain cell types to NO and ONOO- exposure may be dependent on factors such as the intracellular reduced glutathione (GSH) concentration and an ability to increase glycolytic flux in the face of mitochondrial damage. Thus neurones, in contrast to astrocytes, appear particularly vulnerable to the action of these molecules. Following cytokine exposure, astrocytes can increase NO generation, due to de novo synthesis of the inducible form of nitric oxide synthase (NOS). Whilst the NO/ONOO- so formed may not affect astrocyte survival, these molecules may diffuse out to cause mitochondrial damage, and possibly cell death, to other cells, such as neurones, in close proximity. Evidence is now available to support this scenario for neurological disorders, such as multiple sclerosis. In other conditions, such as ischaemia, increased availability of glutamate may lead to an activation of a calcium-dependent nitric oxide synthase associated with neurones. Such increased/inappropriate NO formation may contribute to energy depletion and neuronal cell death. The evidence available for NO/ONOO--mediated mitochondrial damage in various neurological disorders is considered and potential therapeutic strategies are proposed.


Subject(s)
Mitochondria/metabolism , Nervous System Diseases/etiology , Nitric Oxide/metabolism , Amyotrophic Lateral Sclerosis/etiology , Astrocytes/metabolism , Astrocytes/pathology , Cell Death , Electron Transport/genetics , Glutamic Acid/metabolism , Humans , Mitochondria/pathology , Nervous System Diseases/genetics , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/genetics , Neurons/metabolism , Neurons/pathology , Nitrates/metabolism , Permeability
19.
J Neurochem ; 72(4): 1750-8, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10098886

ABSTRACT

Treatment of cultured rat astrocytes with lipopolysaccharide (LPS; 1 microg/ml) increased mRNA expression of glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting step in the pentose phosphate pathway (PPP), in a time-dependent fashion (0-24 h). This effect was accompanied by an increase in G6PD activity (1.74-fold) and in the rate of glucose oxidation through the PPP (6.32-fold). Inhibition of inducible nitric oxide synthase (iNOS) activity by 2-amino-5,6-dihydro-6-methyl-4H-1,3-thiazine (AMT; 50 microM) did not alter the LPS-mediated enhancement of G6PD mRNA expression or PPP activity. Blockade of nuclear factor-kappaB (NF-kappaB) activation by N-benzyloxycarbonyl-Ile-Glu-(O-tert-butyl)-Ala-leucinal (1 microM) prevented the expression of both iNOS mRNA and G6PD mRNA, suggesting that iNOS and G6PD are co-induced by LPS through a common transcriptional pathway involving NF-kappaB activation. Incubation of cells with LPS for 24 h increased intracellular NADPH concentrations (1.63-fold) as compared with untreated cells, but GSH concentrations were not modified by LPS treatment up to 60 h of incubation. However, inhibition of G6PD activity by dehydroepiandrosterone (DHEA; 100 microM), which prevented LPS-mediated enhancements in PPP activity and NADPH concentrations, caused a 50% decrease in the GSH/GSSG ratio after 24-36 h and in GSH concentrations after 60 h of incubation. Furthermore, the changes in glutathione concentrations caused by DHEA were abolished by AMT, suggesting that nitric oxide and/or its reactive derivatives would be involved in this process. From these results, we conclude that LPS-mediated G6PD expression prevents GSH depletion due to nitric oxide and suggest that this phenomenon may be a contributing factor in the defense mechanisms that protect astrocytes against nitric oxide-mediated cell injury.


Subject(s)
Astrocytes/enzymology , Glucosephosphate Dehydrogenase/genetics , Glutathione/metabolism , Nitric Oxide/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Astrocytes/cytology , Blotting, Northern , Cells, Cultured , Dehydroepiandrosterone/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glucosephosphate Dehydrogenase/metabolism , Lipopolysaccharides , NADP/analysis , NF-kappa B/metabolism , Nerve Degeneration/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Pentose Phosphate Pathway/physiology , Prosencephalon/cytology , RNA, Messenger/analysis , Rats , Rats, Wistar , Thiazines/pharmacology
20.
Brain Res ; 816(2): 580-6, 1999 Jan 23.
Article in English | MEDLINE | ID: mdl-9878883

ABSTRACT

Mitochondria have been considered to be a target for glutamate neurotoxicity. The aim of the present work was to investigate the mechanisms leading to glutamate-mediated mitochondrial deenergization, as measured by mitochondrial membrane potential and cell respiration in cultured neurons. Glutamate exposure to cells induced pronounced mitochondrial depolarization associated with an impairment in neuronal respiration, leading to neuronal ATP depletion. These effects were prevented by both the nitric oxide (. NO) synthase inhibitor Nomega-nitro-l-arginine methyl ester and by the N-methyl-d-aspartate glutamate-subtype receptor inhibitor d-(-)-2-amino-5-phosphopentanoate. Our results suggest that glutamate causes ATP depletion by collapsing mitochondrial membrane potential through a.NO-mediated mechanism.


Subject(s)
Cerebral Cortex/drug effects , Glutamic Acid/toxicity , Mitochondria/drug effects , Neurons/drug effects , Neurotoxins/toxicity , Nitric Oxide/physiology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Enzyme Inhibitors/pharmacology , Linear Models , Membrane Potentials/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Neurons/ultrastructure , Nitric Oxide Synthase/antagonists & inhibitors , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...