Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
1.
Toxicol Appl Pharmacol ; 238(1): 11-9, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19361538

ABSTRACT

Cyanide is a potent inhibitor of mitochondrial oxidative metabolism and produces mitochondria-mediated death of dopaminergic neurons and sublethal intoxications that are associated with a Parkinson-like syndrome. Cyanide toxicity is enhanced when mitochondrial uncoupling is stimulated following up-regulation of uncoupling protein-2 (UCP-2). In this study, the role of a pro-survival protein, Bcl-2, in cyanide-mediated cell death was determined in a rat dopaminergic immortalized mesencephalic cell line (N27 cells). Following pharmacological up-regulation of UCP-2 by treatment with Wy14,643, cyanide reduced cellular Bcl-2 expression by increasing proteasomal degradation of the protein. The increased turnover of Bcl-2 was mediated by an increase of oxidative stress following UCP-2 up-regulation. The oxidative stress involved depletion of mitochondrial glutathione (mtGSH) and increased H2O2 generation. Repletion of mtGSH by loading cells with glutathione ethyl ester reduced H2O2 generation and in turn blocked the cyanide-induced decrease of Bcl-2. To determine if UCP-2 mediated the response, RNAi knock down was conducted. The RNAi decreased cyanide-induced depletion of mtGSH, reduced H2O2 accumulation, and inhibited down-regulation of Bcl-2, thus blocking cell death. To confirm the role of Bcl-2 down-regulation in the cell death, it was shown that over-expression of Bcl-2 by cDNA transfection attenuated the enhancement of cyanide toxicity after UCP-2 up-regulation. It was concluded that UCP-2 up-regulation sensitizes cells to cyanide by increasing cellular oxidative stress, leading to an increase of Bcl-2 degradation. Then the reduced Bcl-2 levels sensitize the cells to cyanide-mediated cell death.


Subject(s)
Cell Death/drug effects , Cyanides/toxicity , Ion Channels/drug effects , Mitochondrial Proteins/drug effects , Proto-Oncogene Proteins c-bcl-2/drug effects , Animals , Cell Line, Transformed , DNA, Complementary , Dopamine/metabolism , Down-Regulation/drug effects , Glutathione/drug effects , Glutathione/metabolism , Hydrogen Peroxide/metabolism , Ion Channels/genetics , Mesencephalon/cytology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Pyrimidines/pharmacology , RNA Interference , Rats , Transfection , Uncoupling Protein 2 , Up-Regulation/drug effects
2.
J Neurosci Res ; 86(6): 1397-408, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18183618

ABSTRACT

1Alpha,25-dihydroxyvitamin D(3) (VD(3)) is a neuroprotectant that can reduce cytotoxicity produced by a variety of toxicants. The mechanism of the neuroprotection was studied in rat primary cortical cells in which Wy14,643, an agonist of peroxisome proliferator activated receptor-alpha (PPARalpha), enhances cyanide (KCN) neurotoxicity. In this cell model, Wy14,643 pretreatment enhanced cyanide-induced cell death, and the increased cell death was linked to up-regulation of uncoupling protein-2 (UCP-2). VD(3) reversed cyanide-induced mitochondrial dysfunction in cells pretreated with Wy14,643, as reflected by restoration of cellular ATP and mitochondrial membrane potential (DeltaPsi(m)). Analysis of cellular state 4 oxygen consumption showed increased mitochondrial uncoupling accompanied by up-regulation of UPC-2. The uncoupling was attenuated by prior treatment with VD(3). The interaction of VD(3) with UCP-2 was attributed to increased expression of IkappaB, an inhibitor of NF-kappaB (transcription factor that regulates UCP-2 expression). The increased IkappaB levels lead to reduced nuclear translocation and DNA binding of nuclear factor-kappaB. The role of oxidative stress in the response was then evaluated. Cotreatment with Wy14,643 and cyanide markedly increased reactive oxygen species generation and decreased reduced glutathione levels. The oxidative stress was blocked by VD(3) pretreatment. It was concluded that VD(3) blocks Wy14,643 enhancement of cyanide neurotoxicity by suppressing the redox-mediated transcriptional up-regulation of UCP-2, resulting in reduced mitochondrial proton leak and stabilization of mitochondrial function.


Subject(s)
Cyanides/toxicity , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Vitamin D/analogs & derivatives , Animals , Antioxidants/pharmacology , Blotting, Western , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Glutathione/drug effects , Glutathione/metabolism , Ion Channels/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Proteins/drug effects , Mutagens/toxicity , NF-kappa B/drug effects , NF-kappa B/metabolism , Neurons/metabolism , Neurons/pathology , Oxygen Consumption/drug effects , Pyrimidines/toxicity , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Uncoupling Protein 2 , Up-Regulation , Vitamin D/pharmacology
3.
Free Radic Biol Med ; 43(1): 117-27, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17561100

ABSTRACT

Cyanide produces degeneration of the nervous system in which different modes of cell death are activated in the vulnerable brain areas. In brain, the mechanism underlying the cell death is not clear. In this study, an immortalized dopaminergic cell line was used to characterize the cell death signaling cascade activated by cyanide. Cyanide-treated cells exhibited a time- and concentration-dependent apoptosis that was caspase independent. Cyanide induced a rapid surge of intracellular reactive oxygen species (ROS) generation, followed by p38 mitogen-activated protein kinase (MAPK) activation and nuclear accumulation of hypoxia-inducible factor-1alpha (HIF-1alpha). Activation of p38 MAPK and HIF-1alpha accumulation were attenuated by N-acetyl-L-cysteine (antioxidant), catalase (hydrogen peroxide scavenger), or a selective p38 MAPK inhibitor (SB203580). Cyanide activated the hypoxia response element (HRE) promoter, which was also blocked by the antioxidants and SB203580. HRE activation was followed by increased BNIP3 gene transcription, as reflected by elevated BNIP3 mRNA and protein levels. BNIP3 upregulation was reduced by selective RNAi knockdown of HIF-1alpha. Overexpression of BNIP3 produced mitochondrial dysfunction (reduced membrane potential), caspase-independent apoptosis, and sensitization of the cells to cyanide-induced toxicity. Expression of a dominant-negative mutant or RNAi knockdown of BNIP3 protected the cells from cyanide. It was concluded that cyanide activated the HIF-1alpha-mediated pathway of BNIP3 induction through a redox-sensitive process. Increased BNIP3 expression then served as an initiator of mitochondrial-mediated death.


Subject(s)
Apoptosis , Cyanides/toxicity , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Proteins/metabolism , Mitochondria/physiology , Proto-Oncogene Proteins/metabolism , Acetylcysteine/pharmacology , Animals , Caspases/metabolism , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Imidazoles/pharmacology , Membrane Potential, Mitochondrial , Membrane Proteins/genetics , Mice , Oxidation-Reduction , Oxidative Stress/genetics , Proto-Oncogene Proteins/genetics , Pyridines/pharmacology , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Reactive Oxygen Species/metabolism , Transcription, Genetic , Up-Regulation , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Neuroscience ; 150(1): 159-67, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17980495

ABSTRACT

Bcl-2/adenovirus E1B 19-kDa-interacting protein 3 (BNIP3), a Bcl-2 homology domain 3 (BH3) domain only protein, has been identified as a mitochondrial mediator of hypoxia-induced cell death. Since cyanide produces histotoxic anoxia (chemical hypoxia), the present study was undertaken in primary rat cortical cells to determine involvement of the BNIP3 signaling pathway in cyanide-induced death. Over a 20 h exposure KCN increased BNIP3 expression, followed by a concentration-related apoptotic death. To determine if BNIP3 plays a role in the cell death, expression was either increased with BNIP3 cDNA (BNIP3+) or knocked down with small interfering RNA (RNAi). In BNIP3+ cells, cyanide-induced apoptotic death was markedly enhanced and preceded by reduction of mitochondrial membrane potential (delta psim), release of cytochrome c from mitochondria and elevated caspase 3 and 7 activity. Pretreatment with the pan-caspase inhibitor N-benzyloxycarbonyl-Ala-Asp-fluoromethyl ketone (zVAD-fmk) suppressed BNIP3+-mediated cell death, thus confirming a caspase-dependent apoptosis. On the other hand, BNIP3 knockdown by RNAi or antagonism of BNIP3 by a transmembrane-deleted dominant-negative mutant (BNIP3 delta TM) markedly reduced cell death. Immunohistochemical imaging showed that cyanide stimulated translocation of BNIP3 from cytosol to mitochondria and displacement studies with BNIP3 delta TM showed that integration of BNIP3 into the mitochondrial outer membrane was necessary for the cell death. In BNIP3+ cells, cyclosporin-A, an inhibitor of mitochondrial pore transition, blocked the cyanide-induced reduction of delta psim and decreased the apoptotic death. These results demonstrate in cortical cells that cyanide induces a rapid upregulation of BNIP3 expression, followed by translocation to the mitochondrial outer membrane to reduce delta psim. This was followed by mitochondrial release of cytochrome c to execute a caspase-dependent cell death.


Subject(s)
Apoptosis/drug effects , Cerebral Cortex/cytology , Cyanides/pharmacology , Membrane Proteins/metabolism , Mitochondria/metabolism , Neurons/drug effects , Proto-Oncogene Proteins/metabolism , Analysis of Variance , Animals , Caspases/metabolism , Cells, Cultured , Cyclosporine/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , In Situ Nick-End Labeling , Membrane Potential, Mitochondrial/drug effects , Membrane Proteins/genetics , Mitochondrial Proteins , Mutation/physiology , Protein Transport/drug effects , Protein Transport/physiology , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/pharmacology , Rats , Time Factors , Transfection/methods
5.
Toxicol Sci ; 86(1): 116-24, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15800031

ABSTRACT

Uncoupling protein 2 (UCP-2) is expressed in the inner mitochondrial membrane and modulates mitochondrial function by partially uncoupling oxidative phosphorylation, and it has been reported to modulate cell death. Cyanide is a potent neurotoxin that inhibits complex IV to alter mitochondrial function to induce neuronal death. In primary rat cortical cells KCN produced an apoptotic death at 200-400 microM. Higher concentrations of potassium cyanide (KCN) (500-600 microM) switched the mode of death from apoptosis to necrosis. In necrotic cells, ATP levels were severely depleted as compared to cortical cells undergoing apoptosis. To determine if UCP-2 expression could alter KCN-induced cell death, cells were transiently transfected with full-length human UCP-2 cDNA (UCP-2+). Overexpression switched the mode of death produced by KCN (400 microM) from apoptosis to necrosis. The change in cell death was mediated by impaired mitochondrial function as reflected by a marked decrease of ATP levels and reduction in mitochondrial membrane potential. RNA interference or transfection with a dominant interfering mutant blocked the necrotic response observed in UCP-2+ cells. Additionally, treatment of UCP-2+ cells with cyclosporin A blocked necrosis, indicating the involvement of mitochondrial permeability pore transition in the necrotic death. These results show that increased expression of UCP-2 alters the response to a potent mitochondrial toxin by switching the mode of cell death from apoptosis to necrosis. It is concluded that UCP-2 levels influence cellular responses to cyanide-induced mitochondrial dysfunction.


Subject(s)
Cyanides/toxicity , Mitochondria/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Mitochondria/physiology , Rats , Rats, Sprague-Dawley
6.
Neuropharmacology ; 34(2): 205-10, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7542370

ABSTRACT

In PC12 cells, isosorbide dinitrate (ISDN) and S-nitrosol-acetyl-penicillamine (SNAP), both nitric oxide (NO) generators, attenuated K+ (56 mM)-stimulated release of dopamine. The attenuation was not observed with isosorbide, an ISDN analog lacking NO generating capacity. In this model, A23187 (Ca2+ ionophore), Bay K8644 (Ca2+ slow channel agonist) and veratridine (Na+ channel agonist) stimulated dopamine release. Treatment with ISDN enhanced Bay K8644 and veratridine-evoked dopamine release, while ISDN had no significant effect on the A23187 response. Incubation with 8-bromo-cGMP (membrane permeable cGMP analog) had no effect on basal or stimulated dopamine release in these cells, suggesting NO's response was not mediated by cGMP. In additional studies, K+ (56 mM), Bay K8644 and veratridine elevated cytosolic free calcium levels ([Ca2+]i). ISDN reduced K(+)-stimulated increase in [Ca2+]i, but enhanced the increases of [Ca2+]i induced by Bay K8644 or veratridine. These results suggest NO interacts with K(+)-induced membrane depolarization (possibly by inhibiting membrane conductance to K+) to attenuate Ca2+ influx and Ca(2+)-mediated dopamine secretion stimulated by K+.


Subject(s)
Dopamine/metabolism , Isosorbide Dinitrate/pharmacology , Nitric Oxide/pharmacology , Potassium Compounds/pharmacology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Calcimycin/pharmacology , Calcium/metabolism , Dose-Response Relationship, Drug , PC12 Cells , Rats
7.
Biochem Pharmacol ; 34(16): 2853-8, 1985 Aug 15.
Article in English | MEDLINE | ID: mdl-2992520

ABSTRACT

Anti-idiotypic antibodies which recognize the opiate receptor were generated in guinea pigs following immunization against purified rabbit anti-morphine antibodies. The anti-idiotypic antibodies produced a concentration-dependent inhibition of [3H]naloxone binding to opiate receptors in a membranous mouse brain preparation. Saturation analysis indicated that the antibodies produced a non-competitive inhibition of naloxone binding. The ability of the antibodies to interact with biological systems was investigated in in vitro systems. In both the isolated guinea pig ileal longitudinal muscle and mouse vas deferens, the antibodies produced a concentration-dependent, opiate agonist-like action. The anti-morphine anti-idiotypic antibodies appear to interact specifically with the opiate receptor and may serve as useful tools in characterization of this receptor system.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Immunoglobulin Idiotypes/immunology , Morphine/immunology , Receptors, Opioid/metabolism , Animals , Brain/metabolism , Guinea Pigs , Ileum/metabolism , In Vitro Techniques , Male , Mice , Morphine/metabolism , Naloxone/metabolism , Rabbits , Vas Deferens/metabolism
8.
Biochem Pharmacol ; 41(1): 119-23, 1991 Jan 01.
Article in English | MEDLINE | ID: mdl-1986736

ABSTRACT

Gamma-Aminobutyric acid (GABA) and GABAA agonists enhance stimulated release of [3H]norepinephrine [( 3H]NA) in several regions of the rat brain. In this study, the mechanisms by which GABA and GABAergic agonists augment potassium-stimulated release of [3H]NA from rat frontal cortical slices were examined. GABA enhanced potassium-stimulated [3H]NA release, but did not alter release of [3H]NA evoked by the calcium ionophore A23187, 10(-5) M, either in the presence or the absence of extracellular calcium. The effect of GABA on potassium-stimulated [3H]NA release was apparently reduced by the GABAA antagonist bicuculline methiodide, 10(-4) M, and by the selective inhibitor of GABA uptake SKF 89976A, 10(-5) M, but was abolished only when bicuculline methiodide and SKF 89976A were present in combination. The GABAA agonist muscimol enhanced potassium-stimulated release of [3H]NA in a manner similar to GABA. In addition, nipecotic acid, a substrate for GABA uptake, enhanced potassium-stimulated [3H]NA release. Thus, GABA appears to enhance potassium-stimulated [3H]NA release by acting upon both GABA uptake and GABAA receptors. The GABAA receptors involved in this effect may be a subtype of GABAA receptors since they are not modulated by benzodiazepines. These results support the involvement of the GABA uptake carrier and the GABAA receptor in mediating the enhancement by GABA of potassium-stimulated [3H]NA release in the cortex of the rat.


Subject(s)
Frontal Lobe/drug effects , Norepinephrine/metabolism , Potassium/pharmacology , gamma-Aminobutyric Acid/pharmacology , Animals , Calcimycin/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Frontal Lobe/metabolism , GABA Antagonists , In Vitro Techniques , Male , Muscimol/pharmacology , Rats , Rats, Inbred Strains , Tritium
9.
Biochem Pharmacol ; 42 Suppl: S121-6, 1991 Dec 11.
Article in English | MEDLINE | ID: mdl-1662949

ABSTRACT

Agents that enhance gamma-aminobutyric acid (GABA) neurotransmission can modulate certain effects of opioids, such as analgesia. In this study, the interaction between morphine and GABAergic agents on the release of [3H]norepinephrine ([3H]NE) from rat frontal cortical slices was examined. GABA (10(-4) M), enhanced potassium-stimulated [3H]NE release and reversed the inhibitory effect of 10(-6) M morphine. GABA and muscimol modulated the inhibitory effect of morphine in a noncompetitive manner. Bicuculline methiodide (10(-4) M) reduced the effect of GABA in the absence of morphine, and appeared to reduce the effect of GABA in the presence of morphine, although the latter effect was not statistically significant from the controls. While the GABAA agonist muscimol mimicked the effect of GABA, the GABAB agonist baclofen did not affect the release of [3H]NE in the absence or the presence of 10(-6) M morphine. These results support the involvement of GABAA receptors in modulating the action of opioids on the noradrenergic system in the cerebral cortex of the rat.


Subject(s)
Morphine/pharmacology , Norepinephrine/metabolism , Receptors, GABA-A/drug effects , gamma-Aminobutyric Acid/pharmacology , Animals , Baclofen/pharmacology , Cerebral Cortex/drug effects , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Norepinephrine/antagonists & inhibitors , Rats , Rats, Inbred Strains
10.
Biochem Pharmacol ; 38(19): 3175-8, 1989 Oct 01.
Article in English | MEDLINE | ID: mdl-2554917

ABSTRACT

AC glioma cells, a clonal cell line derived from a rat glioma, responded to 1 mM dibutyryl-cyclic AMP and isobutylmethylxanthine with a change to stellate morphology. A concentration-related morphological change was induced by beta 1- and beta 2-adrenergic agonists with the order of potency being isoproterenol greater than soterenol greater than norepinephrine. Propranolol (nonselective, beta-antagonist), butoxamine (beta 2-antagonist) and metoprolol (beta 1-antagonist) significantly decreased the cell response to isoproternol. Schild analysis of the response, using the competitive antagonist metoprolol, gave pA2 values of 7.5 and 8.5 for the agonists norepinephrine and soterenol, respectively, with slopes of the curves being less than unity. These observations indicate that both beta 1- and beta 2-adrenergic receptors mediate the change in cellular morphology.


Subject(s)
Glioma/analysis , Receptors, Adrenergic, beta/analysis , Animals , Cyclic AMP/physiology , Ethanolamines/pharmacology , Glioma/pathology , Isoproterenol/pharmacology , Metoprolol/pharmacology , Norepinephrine/pharmacology , Rats , Receptors, Adrenergic, beta/drug effects , Tumor Cells, Cultured
11.
Neurosci Res ; 49(1): 13-8, 2004 May.
Article in English | MEDLINE | ID: mdl-15099699

ABSTRACT

Cyanide is generated in neurons and this report examines the two different receptors which mediate cyanide formation in neuronal tissue. An opiate receptor blocked by naloxone increases cyanide production both in rat brain and in rat pheochromocytoma (PC12) cells. A muscarinic receptor in PC12 cells releases cyanide and the effect is blocked by atropine. In rat brain, in vivo, a muscarinic agonist inhibits cyanide generation, possibly by acting on receptor subtypes different from those in PC12 cells. Cyanide generation by a muscarinic agonist in PC12 cells is blocked by pertussis toxin but that caused by an opiate is not. Thus, two different receptors and two different second messenger systems can mediate cyanide generation in PC12 cells. In parallel with the in vivo data, cultured primary rat cortical cells also show decreased cyanide release following muscarinic stimulation. Both blockade of cyanide generation by muscarinic receptor activation and cyanide release by opiate agonists from cortical cells are pertussis toxin insensitive. Similarly, little cyanide generation was seen following cholera toxin treatment. These data indicate that opiate receptors increase and muscarinic receptors decrease cyanide production in rat brain tissue by G-protein independent mechanisms. This work supports the suggestion that the powerful actions of cyanide may be important for neuromodulation in the CNS.


Subject(s)
Brain/metabolism , Cyanides/metabolism , Oxotremorine/analogs & derivatives , PC12 Cells/metabolism , Receptors, Cell Surface/metabolism , Analysis of Variance , Animals , Atropine/pharmacology , Brain/drug effects , Carbachol/pharmacology , Cells, Cultured , Cholinergic Agonists/pharmacology , Drug Interactions , Embryo, Mammalian , Hydromorphone/pharmacology , Morphine/pharmacology , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Narcotics/pharmacology , Oxotremorine/pharmacology , PC12 Cells/drug effects , Pertussis Toxin/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/drug effects
12.
Toxicol Sci ; 64(1): 83-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11606804

ABSTRACT

In evaluating mechanisms of trimethyltin (TMT)-initiated neuronal damage, the present study focused on involvement of reactive oxygen species, protein kinase C (PKC), and glutamate receptors. Exposure of cerebellar granule cells to TMT (0.01-0.1 microM) produced primarily apoptosis, but higher concentrations were associated with cellular lactate dehydrogenase efflux and necrosis. TMT increased generation of cellular reactive oxygen species, which was inhibited by either L-NAME (inhibitor of nitric oxide synthase, NOS) or catalase, indicating that both NO and H(2)O(2) are formed on TMT exposure. Since chelerythrine (selective PKC inhibitor) also inhibited oxidative species generation, PKC appears to play a significant role in TMT-induced oxidative stress. The metabotropic glutamate receptor antagonist, MCPG, (but not MK-801) prevented oxidative species generation, indicating significant involvement of metabotropic receptors (but not NMDA receptors) in TMT-induced oxidative stress. NOS involvement in the action of TMT was confirmed through measurement of nitrite, which increased concentration dependently. Nitrite accumulation was blocked by L-NAME, chelerythrine, or MCPG, showing that NO is generated by TMT and that associated changes in NOS are regulated by a PKC-mediated mechanism. Oxidative damage by TMT was demonstrated by detection of elevated malondialdehyde levels. It was concluded that low concentrations of TMT (0.01-0.1 microM) cause apoptotic cell death in which oxidative signaling is an important event. Higher concentrations of TMT initiate necrotic death, which involves both an oxidative and a non-oxidative component. TMT-induced necrosis but not apoptosis in granule cells is mediated by glutamate receptors.


Subject(s)
Apoptosis/drug effects , Cerebellum/drug effects , Necrosis , Neurons/drug effects , Trimethyltin Compounds/toxicity , Alkaloids , Animals , Apoptosis/physiology , Benzophenanthridines , Catalase/pharmacology , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Dizocilpine Maleate/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , In Situ Nick-End Labeling , L-Lactate Dehydrogenase/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Neurons/cytology , Neurons/metabolism , Nitrites/metabolism , Phenanthridines/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Reactive Oxygen Species/metabolism , Receptors, Glutamate/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Thiobarbituric Acid Reactive Substances/metabolism , Trimethyltin Compounds/administration & dosage
13.
Toxicol Sci ; 75(1): 99-107, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12805646

ABSTRACT

Execution of cyanide-induced apoptosis is mediated by release of cytochrome c from mitochondria. To determine how cyanide initiates cytochrome c release, Bax translocation was investigated in primary cultures of cortical neurons. Under nonapoptotic (control) conditions, Bax resided predominantly in the cytoplasm. After 300-microM cyanide treatment for 1 h, Bax translocated to the mitochondria, as shown by immunocytochemical staining and subcellular fractionation; Western blot analysis confirmed "cytosol-to-mitochondria" translocation of Bax. Temporal analysis showed that Bax translocation preceded cytochrome c release from the mitochondria, which was initiated 3 h after cyanide treatment. In double-immunofluorescence labeling for both Bax and cytochrome c, it was observed that cytochrome c was released only in cells showing Bax in mitochondria. The role of p38 mitogen-activated protein (MAP) kinase in Bax translocation was studied. The p38 MAP kinase was activated 30 min after cyanide, and its phosphorylation level of activity began to decrease 3 h later. SB203580, a p38 MAP kinase inhibitor, blocked translocation of Bax to mitochondria, whereas SB202474, a control peptide, had no effect on translocation. Inhibition of p38 MAP kinase by SB203580 blocked all downstream effects of Bax translocation, including cytochrome c release, caspase activation, and internucleosomal DNA fragmentation. These results demonstrated that Bax translocation is critical for cyanide-induced cytochrome c release and that p38 MAP kinase regulates Bax translocation from cytosol to mitochondria.


Subject(s)
Apoptosis/drug effects , Mitogen-Activated Protein Kinases/metabolism , Potassium Cyanide/toxicity , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Animals , Blotting, Western , Caspases/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Cytochromes c/biosynthesis , Cytoplasm/metabolism , Enzyme Inhibitors/pharmacology , Mitochondria/metabolism , Neurons/metabolism , Neurons/ultrastructure , Protein Transport , Rats , Rats, Sprague-Dawley , bcl-2-Associated X Protein , p38 Mitogen-Activated Protein Kinases
14.
J Neurosci Methods ; 61(1-2): 15-21, 1995.
Article in English | MEDLINE | ID: mdl-8618413

ABSTRACT

A method for rapid fluorometric assay of intracellular nitric oxide (NO) formation was developed for use in cultured neuronal cells. In a cell-free system 2,7-dichlorofluorescin (DCF), a non-fluorescent species, is oxidized by NO to dichlorofluorescein, a fluorescent compound. Addition of NO to a solution containing DCF increased the fluorescent signal within 10 s and continued to increase slowly over a 10-min period. The intensity of the fluorescence was dependent upon the concentration of NO. In DCF-loaded PC12 cells, addition of NO markedly increased fluorescence (limit of detection = 16 microM NO) and pretreatment with reduced hemoglobin (Hb) inhibited the NO-mediated increase of fluorescence in both the cell-free system and PC12 cells. In PC12 cells loaded with DCF, the NO generator sodium nitroprusside (SNP) produced a rapid increase of fluorescence. To rule out the possibility that reactive oxygen species (ROS) mediated the increased of fluorescence, superoxide dismutase (SOD) and catalase were added to the cuvette. The enzymes did not alter the fluorescence generated after addition of NO to PC12 cells. This assay was used to determine the ability of glutamate to stimulate NO production in cerebellar granule cells. When 10 microM glutamate was added to DCF-loaded cerebellar granule cells, a rapid increase in fluorescence was noted. The fluorescence was blocked approximately 50% after addition of either Hb or SOD, or by pretreatment with NG-nitro-L-arginine methyl ester (300 microM), a nitric oxide synthase (NOS) inhibitor. It was concluded that glutamate stimulated intracellular generation of both NO and ROS, and at least 50% of the oxidation of DCF was attributed to intracellular generation of NO. These results demonstrate that oxidation of DCF by NO can be used to measure intracellular generation of NO and by adding either Hb or SOD to the cell system, the extent of oxidation of DCF attributed to NO and ROS can be determined.


Subject(s)
Fluoresceins , Nitric Oxide/biosynthesis , PC12 Cells/metabolism , Analysis of Variance , Animals , Cells, Cultured/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Rats
15.
Brain Res ; 676(1): 205-11, 1995 Apr 03.
Article in English | MEDLINE | ID: mdl-7796171

ABSTRACT

The effect of potassium cyanide-induced chemical hypoxia on protein kinase C (PKC) translocation and cell injury was studied in differentiated PC12 cells. The cellular distribution of PKC in control cells and cells exposed to 100 microM and 1 mM KCN for 30 min. was visualized by use of an anti-PKC antibody and confocal laser scanning microscope. In control differentiated PC12 cells, PKC was localized perinuclearly, while following 12-phorbol 13-myristate acetate (PMA) or KCN it was translocated to the plasma and organelle membranes. Western blot analysis was used to quantify the translocation. Chemical hypoxia increased the membrane-bound PKC to 210% of control levels, while chelerythrine, a PKC inhibitor, and block of calcium influx into the cells (with calcium channel blocker and calcium-free medium) prevented this effect. Cyanide-induced PKC translocation persisted for at least 120 min. Cell injury was monitored by measuring lactate dehydrogenase (LDH) efflux from the cells 24 hr after addition of cyanide. PKC activation plays a role in hypoxic damage, since PKC down-regulation (by overnight exposure to PMA) or inhibition (with chelerythrine or staurosporine) conferred protection against KCN-induced cytotoxicity. Ca2+ channel blocker nifedipine also protected against chemical hypoxia. None of the pretreatments rendered complete protection against cyanide-induced hypoxia, indicating that PKC-independent mechanism(s) are also activated during chemical hypoxia and contribute to cell injury.


Subject(s)
Cell Hypoxia/drug effects , Protein Kinase C/metabolism , Alkaloids , Animals , Benzophenanthridines , Blotting, Western , Cell Survival/drug effects , Immunohistochemistry , Microscopy, Confocal , Nifedipine/pharmacology , PC12 Cells/metabolism , Phenanthridines/pharmacology , Potassium Cyanide/pharmacology , Rats
16.
Brain Res ; 593(1): 114-6, 1992 Oct 09.
Article in English | MEDLINE | ID: mdl-1458312

ABSTRACT

Exposure of primary hippocampal cultures to NaCN (2 mM) or glyburide (5 microM) alone for 3 h did not produce a rise in extracellular lactic dehydrogenase (LDH) activity. Coincubation with NaCN and glyburide produced a significant efflux of LDH from the neurons. Diazoxide or D-2-amino-5-phosphovalerate (APV) partially reversed the release of LDH by the combination of NaCN and glyburide. These observations indicate ATP-sensitive potassium channels (KATP) are activated by nonlethal concentrations of cyanide and their blockade with glyburide unmasks cyanide's toxicity. The cytotoxicity of cyanide appears to result from a combination of processes resulting in altered ion handling and excitotoxicity.


Subject(s)
Adenosine Triphosphate/pharmacology , Diazoxide/toxicity , Glyburide/toxicity , Hippocampus/drug effects , Neurons/drug effects , Neurotoxins/toxicity , Potassium Channels/drug effects , Sodium Cyanide/toxicity , Animals , Cells, Cultured , Drug Synergism , Hippocampus/pathology , Kinetics , L-Lactate Dehydrogenase/metabolism , Neurons/pathology , Neurons/physiology , Potassium Channels/physiology , Valine/analogs & derivatives , Valine/pharmacology
17.
Brain Res ; 768(1-2): 294-300, 1997 Sep 12.
Article in English | MEDLINE | ID: mdl-9369328

ABSTRACT

Hydrogen cyanide, a gaseous molecule, is produced by white blood cells during phagocytosis. The present study examined the possibility that neuronal-like cells may also produce cyanide following activation. Rat pheochromocytoma (PC12) cells exhibited a low level of cyanide generation that was significantly increased by mu-opiate agonists (hydromorphone, morphine) and blocked by naloxone. A variety of other agonists including bradykinin, nicotine and glutamate did not generate cyanide in PC12 cells. Systemic administration of hydromorphone to rats increased brain cyanide levels by 61% after 15 min. Using microdialysis probes implanted in the cortical-hippocampal areas of the anesthetized rat or in the hypothalamus of the conscious hamster, a 2- to 5-fold increase in cyanide generation was seen after hydromorphone administration and this increase was blocked by naloxone. To determine whether cyanide release by hydromorphone has functional significance in a neuronal system, cyanide enhancement of N-methyl-D-aspartate (NMDA)-induced increased [Ca2+]i was measured in rat cerebellar granule cells. Hydromorphone enhanced the response to NMDA similar to cyanide and the hydromorphone effect was blocked by cyanide scavengers. These data show that cyanide generation is increased in neuronal tissue by a mu-opiate receptor agonist and it is proposed that endogenous cyanide may modulate the NMDA receptor response.


Subject(s)
Hydrogen Cyanide/pharmacology , Neurotransmitter Agents/pharmacology , Receptors, Opioid, mu/agonists , Analgesics, Opioid/pharmacology , Animals , Cells, Cultured , Cricetinae , Female , Hydrogen Cyanide/metabolism , Hydromorphone/pharmacology , In Vitro Techniques , N-Methylaspartate/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neurons/drug effects , Neurons/metabolism , Neurotransmitter Agents/metabolism , PC12 Cells , Rats , Rats, Sprague-Dawley
18.
Eur J Pharmacol ; 102(1): 187-90, 1984 Jun 15.
Article in English | MEDLINE | ID: mdl-6090156

ABSTRACT

Antibodies that specifically recognize and bind opiate receptors were developed by the anti-idiotypic method. Antimorphine antibodies were generated in rabbits and showed selective binding to various opiate ligands. Guinea pigs were immunized with antimorphine antibodies to produce anti-idiotypic antibodies that cross-reacted with opiate receptors. Guinea pig antiserum inhibited binding of naloxone (0.6 nmol) to mouse brain homogenate. In isolated mouse vas deferens, contraction of electrically stimulated tissue was inhibited by the antiserum. The results indicate that anti-idiotypic antibodies which interact with opiate receptors were developed without isolating receptors.


Subject(s)
Immunoglobulin Idiotypes/immunology , Morphine/immunology , Receptors, Opioid/immunology , Animals , Guinea Pigs , In Vitro Techniques , Male , Mice , Muscle, Smooth/metabolism , Naloxone/metabolism , Rabbits , Vas Deferens/metabolism
19.
Eur J Pharmacol ; 213(3): 317-21, 1992 Mar 31.
Article in English | MEDLINE | ID: mdl-1377629

ABSTRACT

The ability of atrial natriuretic peptide (ANP) to modulate K+-stimulated release of [3H]norepinephrine ([3H]NE) from rat hypothalamic slices was investigated. ANP-(1-28) significantly decreased K+-stimulated [3H]NE release in a concentration-dependent manner (maximal inhibition = 22% of control with 100 nM, ED50 = 70 pM). Pretreatment with pertussis toxin did not alter the response to ANP. 8Br-cGMP (10 microM), a cGMP analog, significantly decreased [3H]NE release and when combined with 10 nM ANP-(1-28), an additive effect was observed. Additionally, 3-isobutyl 1-methylxanthine (IBMX) (200 microM), a phosphodiesterase inhibitor, combined with ANP-(1-28) 10 nM, significantly decreased [3H]NE release. These results indicate that ANP-(1-28) modulated release of [3H]NE from rat hypothalamic slices and the effect is most likely mediated by elevation of intraneuronal cGMP.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Cyclic GMP/physiology , Hypothalamus/drug effects , Norepinephrine/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Hypothalamus/anatomy & histology , Hypothalamus/metabolism , In Vitro Techniques , Kinetics , Male , Potassium/pharmacology , Rats , Rats, Inbred Strains , Receptors, Atrial Natriuretic Factor , Receptors, Cell Surface/drug effects , Receptors, Cell Surface/physiology , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Stimulation, Chemical , Tritium
20.
Eur J Pharmacol ; 70(2): 121-8, 1981 Mar 12.
Article in English | MEDLINE | ID: mdl-6790294

ABSTRACT

The effects of central administration of (D-Ala2)methionine-enkephalinamide (EA) on respiration were studied in the unanesthetized rat. EA induced a dose-dependent depression of respiratory rate and tidal volume which was reversed by the subcutaneous administration of naloxone HCl (10 mg/kg). Increasing atmospheric CO2 concentration stimulated EA-induced respiratory depression and the peptide produced a parallel shift to the right of both the respiratory rate -- PaCO2 and tidal volume -- PaCO2 curves. Following chronic administration of EA tolerance to the respiratory depression was observed and chronic exposure to morphine produced partial tolerance to the respiratory depression induced by EA. It was concluded that EA desensitizes the central chemoreceptors to CO2 and the action of EA on respiration parallels that of morphine.


Subject(s)
Endorphins/pharmacology , Enkephalin, Methionine/analogs & derivatives , Enkephalins/pharmacology , Respiration/drug effects , Animals , Carbon Dioxide/pharmacology , Dose-Response Relationship, Drug , Drug Tolerance , Male , Naloxone/pharmacology , Rats , Respiratory Center/physiology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL