136 results on '"Isom GE"'
Search Results
2. MECHANISM OF CHLORPROMAZINE ANTAGONISM OF CYANIDE INTOXICATION
- Author
-
Burrows Ge, Isom Ge, James L. Way, and Dodson Ra
- Subjects
chemistry.chemical_compound ,Chemistry ,Cyanide ,medicine ,Pharmacology ,Antagonism ,Chlorpromazine ,Mechanism (sociology) ,medicine.drug - Published
- 1977
- Full Text
- View/download PDF
3. Nitrite-mediated antagonism of cyanide inhibition of cytochrome c oxidase in dopamine neurons.
- Author
-
Leavesley HB, Li L, Mukhopadhyay S, Borowitz JL, and Isom GE
- Subjects
- Animals, Cell Line, Transformed, Cyclic N-Oxides pharmacology, Drug Antagonism, Electron Transport Complex IV metabolism, Free Radical Scavengers pharmacology, Hydrogen Cyanide metabolism, Imidazoles pharmacology, Neurons metabolism, Nitric Oxide metabolism, Rats, Chemical Warfare Agents toxicity, Dopamine physiology, Electron Transport Complex IV antagonists & inhibitors, Hydrogen Cyanide toxicity, Neurons drug effects, Nitric Oxide Donors pharmacology, Sodium Nitrite pharmacology
- Abstract
Cyanide inhibits aerobic metabolism by binding to the binuclear heme center of cytochrome c oxidase (CcOX). Amyl nitrite and sodium nitrite (NaNO(2)) antagonize cyanide toxicity in part by oxidizing hemoglobin to methemoglobin (mHb), which then scavenges cyanide. mHb generation is thought to be a primary mechanism by which the NO(2)(-) ion antagonizes cyanide. On the other hand, NO(2)(-) can undergo biotransformation to generate nitric oxide (NO), which may then directly antagonize cyanide inhibition of CcOX. In this study, nitrite-mediated antagonism of cyanide inhibition of oxidative phosphorylation was examined in rat dopaminergic N27 cells. NaNO(2) produced a time- and concentration-dependent increase in whole-cell and mitochondrial levels of NO. The NO scavenger 2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxy 3-oxide (PTIO) reversed this increase in cellular and mitochondrial NO. NO generated from NaNO(2) decreased cellular oxygen consumption and inhibited CcOX activity. PTIO reversed the NO-mediated inhibition, thus providing strong evidence that NO mediates the action of NaNO(2). Under similar conditions, KCN (20muM) inhibited cellular state-3 oxygen consumption and CcOX activity. Pretreatment with NaNO(2) reversed KCN-mediated inhibition of both oxygen consumption and CcOX activity. The NaNO(2) antagonism of cyanide was blocked by pretreatment with the NO scavenger PTIO. It was concluded that NaNO(2) antagonizes cyanide inhibition of CcOX by generating of NO, which then interacts directly with the binding of KCN x CcOX to reverse the toxicity. In vivo antagonism of cyanide by NO(2)(-) appears to be due to both generation of mHb and direct displacement of cyanide from CcOX by NO.
- Published
- 2010
- Full Text
- View/download PDF
4. Cyanide-induced apoptosis of dopaminergic cells is promoted by BNIP3 and Bax modulation of endoplasmic reticulum-mitochondrial Ca2+ levels.
- Author
-
Zhang L, Li L, Leavesley HW, Zhang X, Borowitz JL, and Isom GE
- Subjects
- Animals, Brain metabolism, Brain pathology, Cell Line, Endoplasmic Reticulum metabolism, Male, Membrane Potential, Mitochondrial drug effects, Membrane Proteins antagonists & inhibitors, Mice, Mitochondria metabolism, Mitochondrial Proteins antagonists & inhibitors, RNA, Small Interfering pharmacology, Up-Regulation, Apoptosis drug effects, Brain drug effects, Calcium metabolism, Dopamine metabolism, Endoplasmic Reticulum drug effects, Membrane Proteins biosynthesis, Mitochondria drug effects, Mitochondrial Proteins biosynthesis, Potassium Cyanide toxicity, bcl-2-Associated X Protein metabolism
- Abstract
Cyanide is a potent neurotoxicant that can produce dopaminergic neuronal death in the substantia nigra and is associated with a Parkinson-like syndrome. In this study involvement of Bcl-2/adenovirus E1B 19-kDa interacting protein 3 (BNIP3), a BH3-only Bcl-2 protein, in cyanide-induced death of dopaminergic cells was determined in mice and Mes 23.5 cells. Treatment of mice with cyanide up-regulated BNIP3 and Bax expression in tyrosine hydroxylase (TH)-positive cells of the substantia nigra, and progressive loss of TH-positive neurons was observed over a 9-day period. In Mes 23.5 dopaminergic cells, cyanide stimulated translocalization of BNIP3 to both endoplasmic reticulum (ER) and mitochondria. In ER, BNIP3 stimulated release of Ca(2+) into the cytosol, followed by accumulation of mitochondrial Ca(2+), resulting in reduction of mitochondrial membrane potential (Deltapsi(m)) and eventually cell death. Cyanide also activated Bax to colocalize with BNIP3 in ER and mitochondria. Forced overexpression of BNIP3 activated Bax, whereas gene silencing reduced Bax activity. Knockdown of Bax expression by small interfering RNA blocked the BNIP3-mediated changes in ER and mitochondrial Ca(2+) to block cyanide-induced mitochondrial dysfunction and cell death. These findings show that BNIP3-mediates cyanide-induced dopaminergic cell death through a Bax downstream signal that mobilizes ER Ca(2+) stores, followed by mitochondrial Ca(2+) overload.
- Published
- 2010
- Full Text
- View/download PDF
5. BNIP3 mediates cell death by different pathways following localization to endoplasmic reticulum and mitochondrion.
- Author
-
Zhang L, Li L, Liu H, Borowitz JL, and Isom GE
- Subjects
- Animals, Cell Line, Tumor, Humans, Membrane Proteins genetics, Mice, Proto-Oncogene Proteins genetics, Rats, Apoptosis, Calcium metabolism, Calcium Signaling, Endoplasmic Reticulum metabolism, Membrane Proteins metabolism, Mitochondria metabolism, Proto-Oncogene Proteins metabolism
- Abstract
BNIP3 (Bcl-2/adenovirus E1B 19-kDa interacting protein 3) is a BH3-only proapoptotic member of the Bcl-2 family. Because the interaction of Bcl-2 proteins with intracellular Ca(2+) stores has been linked to apoptosis, the role of Ca(2+) transfer between endoplasmic reticulum (ER) and mitochondria in BNIP3-mediated cell death was determined in a rat dopaminergic neuronal cell line, Mes 23.5. BNIP3 mutants were constructed to target either ER or mitochondria. Localization of BNIP3 to the ER membrane facilitated release of Ca(2+) and subsequently increased uptake of Ca(2+) into mitochondria. Excessive accumulation of mitochondrial Ca(2+) decreased mitochondrial membrane potential (DeltaPsi(m)), resulting in execution of a caspase-independent cell death. Reduction of ER Ca(2+) induced by ER-targeted BNIP3 and the subsequent cell death was blocked by the antiapoptotic protein, Bcl-2. On the other hand, mitochondria-targeted BNIP3 initiated apoptosis by a Ca(2+)-independent mechanism by inducing mitochondrial pore transition and dissipation of DeltaPsi(m). The disruption of DeltaPsi(m) and cell death was not blocked by Bcl-2 overexpression. These findings show that BNIP3 undergoes a dual subcellular localization and initiates different cell death signaling events in the ER and mitochondria. Bcl-2 counters the BNIP3-initiated mobilization of ER Ca(2+) depletion to reduce the level of apoptosis.
- Published
- 2009
- Full Text
- View/download PDF
6. Cyanide-induced death of dopaminergic cells is mediated by uncoupling protein-2 up-regulation and reduced Bcl-2 expression.
- Author
-
Zhang X, Li L, Zhang L, Borowitz JL, and Isom GE
- Subjects
- 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, Cell Death drug effects, Cyanides toxicity, Ion Channels drug effects, Mitochondrial Proteins drug effects, Proto-Oncogene Proteins c-bcl-2 drug effects
- 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.
- Published
- 2009
- Full Text
- View/download PDF
7. Nicotine and type 2 diabetes.
- Author
-
Borowitz JL and Isom GE
- Subjects
- Adult, Animals, Diabetes Mellitus, Type 2 physiopathology, Disease Models, Animal, Female, Humans, Lactation Disorders etiology, Lactation Disorders physiopathology, Maternal Exposure adverse effects, Pregnancy, Prenatal Exposure Delayed Effects, Diabetes Mellitus, Type 2 etiology, Nicotine adverse effects, Smoking adverse effects, Tobacco Use Disorder
- Published
- 2008
- Full Text
- View/download PDF
8. 1Alpha,25-dihydroxyvitamin D3 attenuates cyanide-induced neurotoxicity by inhibiting uncoupling protein-2 up-regulation.
- Author
-
Li L, Prabhakaran K, Zhang X, Zhang L, Liu H, Borowitz JL, and Isom GE
- Subjects
- 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, Cyanides toxicity, Ion Channels metabolism, Mitochondrial Proteins metabolism, Neurons drug effects, Neuroprotective Agents pharmacology, Vitamin D analogs & derivatives
- 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., (2008 Wiley-Liss, Inc.)
- Published
- 2008
- Full Text
- View/download PDF
9. Interaction of cyanide and nitric oxide with cytochrome c oxidase: implications for acute cyanide toxicity.
- Author
-
Leavesley HB, Li L, Prabhakaran K, Borowitz JL, and Isom GE
- Subjects
- Animals, Cell Line, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, In Situ Hybridization, Kinetics, Laser Scanning Cytometry, Mesencephalon pathology, Mitochondria drug effects, Mitochondria enzymology, Mitochondria metabolism, Nitric Oxide Donors pharmacology, Nitric Oxide Synthase antagonists & inhibitors, Oxygen Consumption drug effects, Rats, S-Nitroso-N-Acetylpenicillamine pharmacology, omega-N-Methylarginine pharmacology, Cyanides toxicity, Electron Transport Complex IV physiology, Nitric Oxide physiology
- Abstract
Acute cyanide toxicity is attributed to inhibition of cytochrome c oxidase (CcOX), the oxygen-reducing component of mitochondrial electron transport; however, the mitochondrial action of cyanide is complex and not completely understood. State-3 oxygen consumption and CcOX activity were studied in rat N27 mesencephalic cells to examine the functional interaction of cyanide and nitric oxide (NO). KCN produced a concentration-dependent inhibition of cellular respiration. Cyanide's median inhibitory concentration (IC50) of oxygen consumption (13.2 +/- 1.8microM) was higher than the CcOX IC50 (7.2 +/- 0.1microM). Based on respiratory threshold analysis, 60% inhibition of CcOX was necessary before oxygen consumption was decreased. Addition of high levels of exogenous NO (100microM S-nitroso-N-acetyl-DL-penicillamine) attenuated cyanide inhibition of both respiration and CcOX. On the other hand, when endogenous NO generation was blocked by an NOS inhibitor (N(omega)-monomethyl-L-arginine ester), the cyanide IC50 for both respiration and CcOX increased to 59.6 +/- 0.9microM and 102 +/- 10microM, respectively, thus showing constitutive, low-level NO production enhanced cyanide inhibition. Laser scanning cytometry showed that cyanide elevated mitochondrial NO, which then was available to interact with CcOX to enhance the inhibition. It is concluded that the rapid, potent action of cyanide is due in part to mitochondrial generation of NO, which enhances inhibition of CcOX. At low mitochondrial oxygen tensions, the cyanide-NO interaction would be increased. Also, the antidotal action of sodium nitrite is partly explained by generation of high mitochondrial levels of NO, which antagonizes the CcOX inhibition.
- Published
- 2008
- Full Text
- View/download PDF
10. Upregulation of BNIP3 and translocation to mitochondria mediates cyanide-induced apoptosis in cortical cells.
- Author
-
Prabhakaran K, Li L, Zhang L, Borowitz JL, and Isom GE
- Subjects
- 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, Apoptosis drug effects, Cerebral Cortex cytology, Cyanides pharmacology, Membrane Proteins metabolism, Mitochondria metabolism, Neurons drug effects, Proto-Oncogene Proteins metabolism
- 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.
- Published
- 2007
- Full Text
- View/download PDF
11. Uncoupling protein-2 up-regulation and enhanced cyanide toxicity are mediated by PPARalpha activation and oxidative stress.
- Author
-
Zhang X, Li L, Prabhakaran K, Zhang L, Leavesley HB, Borowitz JL, and Isom GE
- Subjects
- Animals, Cell Line, Transformed, Cell Survival drug effects, Dose-Response Relationship, Drug, Drug Combinations, Drug Interactions, Gene Silencing, Glutathione analogs & derivatives, Glutathione pharmacology, Indoles pharmacology, Ion Channels genetics, Mitochondria drug effects, Mitochondria metabolism, Mitochondrial Proteins genetics, Neurons drug effects, Neurons metabolism, Neurons pathology, PPAR alpha agonists, Pyrimidines pharmacology, Rats, Reactive Oxygen Species metabolism, Uncoupling Protein 2, Up-Regulation drug effects, Vitamin E pharmacology, Enzyme Inhibitors toxicity, Ion Channels metabolism, Mitochondrial Proteins metabolism, Oxidative Stress, PPAR alpha metabolism, Potassium Cyanide toxicity, Up-Regulation physiology
- Abstract
Uncoupling protein 2 (UCP-2) is an inner mitochondrial membrane proton carrier that modulates mitochondrial membrane potential (DeltaPsi(m)) and uncouples oxidative phosphorylation. We have shown that up-regulation of UCP-2 by Wy14,643, a selective peroxisome proliferator-activated receptor-alpha (PPARalpha) agonist, enhances cyanide cytotoxicity. The pathway by which Wy14,643 up-regulates UCP-2 was determined in a dopaminergic cell line (N27 cells). Since dopaminergic mesencephalic cells are a primary brain target of cyanide, the N27 immortalized mesencephalic cell was used in this study. Wy14,643 produced a concentration- and time-dependent up-regulation of UCP-2 that was linked to enhanced cyanide-induced cell death. MK886 (PPARalpha antagonist) or PPARalpha knock-down by RNA interference (RNAi) inhibited PPARalpha activity as shown by the peroxisome proliferator response element-luciferase reporter assay, but only partially decreased up-regulation of UCP-2. The role of oxidative stress as an alternative pathway to UCP-2 up-regulation was determined. Wy14,643 induced a rapid surge of ROS generation and loading cells with glutathione ethyl ester (GSH-EE) or pre-treatment with vitamin E attenuated up-regulation of UCP-2. On the other hand, RNAi knockdown of PPARalpha did not alter ROS generation, suggesting a PPARalpha-independent component to the response. Co-treatment with PPARalpha-RNAi and GSH-EE blocked both the up-regulation of UCP-2 by Wy14,643 and the cyanide-induced cell death. It was concluded that a PPARalpha-mediated pathway and an oxidative stress pathway independent of PPARalpha mediate the up-regulation of UCP-2 and subsequent increased vulnerability to cyanide-induced cytotoxicity.
- Published
- 2007
- Full Text
- View/download PDF
12. HIF-1alpha activation by a redox-sensitive pathway mediates cyanide-induced BNIP3 upregulation and mitochondrial-dependent cell death.
- Author
-
Zhang L, Li L, Liu H, Prabhakaran K, Zhang X, Borowitz JL, and Isom GE
- Subjects
- 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, Apoptosis, Cyanides toxicity, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Membrane Proteins metabolism, Mitochondria physiology, Proto-Oncogene Proteins metabolism
- 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.
- Published
- 2007
- Full Text
- View/download PDF
13. Inducible nitric oxide synthase up-regulation and mitochondrial glutathione depletion mediate cyanide-induced necrosis in mesencephalic cells.
- Author
-
Prabhakaran K, Li L, Borowitz JL, and Isom GE
- Subjects
- Analysis of Variance, Animals, Blotting, Western methods, Cell Death drug effects, Cells, Cultured, Drug Interactions, Embryo, Mammalian, Enzyme Inhibitors pharmacology, Immunohistochemistry methods, Mitochondria enzymology, Necrosis chemically induced, Neurons enzymology, Neurons pathology, RNA, Messenger biosynthesis, Rats, Rats, Sprague-Dawley, Reverse Transcriptase Polymerase Chain Reaction methods, Thiobarbituric Acid Reactive Substances metabolism, Cyanides pharmacology, Glutathione metabolism, Mesencephalon cytology, Mitochondria drug effects, Neurons drug effects, Nitric Oxide Synthase Type II metabolism, Up-Regulation drug effects
- Abstract
We have previously shown in rat primary cultured mesencephalic cells that cyanide induces a high level of oxidative stress and necrotic death. To evaluate the mechanism of the cytotoxicity, the effects of cyanide on intracellular glutathione (GSH) pools and inducible nitric oxide synthase (iNOS)-mediated reactive nitrogen species (RNS) generation were studied. Cyanide rapidly depleted intracellular GSH. Restoration of GSH blocked cell death, whereas depletion of GSH by synthesis inhibition increased the necrosis. Selective depletion of mitochondrial GSH (mtGSH) increased oxidative stress and enhanced cell death, whereas the cytoplasmic pool was not critical to cell survival. These actions were accompanied by increased iNOS expression as determined by Western blot analysis, RT-PCR and immunohistochemistry. Up-regulation of iNOS led to increased generation of NO as reflected by elevated nitrite levels (an end product of NO metabolism). It was determined by use of a selective inhibitor that up-regulation of iNOS expression was transcriptionally regulated by activation of nuclear factor-kappaB, a redox-sensitive transcription factor. It was concluded that, in cyanide-mediated neurotoxicity, mtGSH is a vital component of the cellular antioxidant defense, and its depletion can lead to oxidative stress-mediated iNOS up-regulation, thus enhancing RNS generation and necrosis., (Copyright 2006 Wiley-Liss, Inc.)
- Published
- 2006
- Full Text
- View/download PDF
14. Trimethyltin-induced apoptosis is associated with upregulation of inducible nitric oxide synthase and Bax in a hippocampal cell line.
- Author
-
Zhang L, Li L, Prabhakaran K, Borowitz JL, and Isom GE
- Subjects
- Amino Acid Chloromethyl Ketones pharmacology, Animals, Caspase 3 metabolism, Caspase Inhibitors, Cell Line, Cyclic N-Oxides pharmacology, Cyclosporine pharmacology, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, Hippocampus cytology, Hippocampus drug effects, Hippocampus metabolism, Membrane Potentials drug effects, Mitochondrial Membrane Transport Proteins antagonists & inhibitors, Mitochondrial Membranes drug effects, Mitochondrial Membranes physiology, Mitochondrial Permeability Transition Pore, NG-Nitroarginine Methyl Ester pharmacology, Nitric Oxide antagonists & inhibitors, Nitric Oxide metabolism, Nitric Oxide Synthase Type II antagonists & inhibitors, Nitrogen Oxides pharmacology, Oligonucleotides, Antisense pharmacology, Peptides pharmacology, Reactive Oxygen Species antagonists & inhibitors, Reactive Oxygen Species metabolism, Trimethyltin Compounds antagonists & inhibitors, Up-Regulation drug effects, Apoptosis drug effects, Nitric Oxide Synthase Type II metabolism, Trimethyltin Compounds toxicity, bcl-2-Associated X Protein metabolism
- Abstract
Trimethyltin (TMT) produces selective neuronal degeneration in the central nervous system (CNS), in which the hippocampus is the most sensitive area. Since previous studies have been conducted in either non-neural cells or mixed primary cultures, an immortalized hippocampal neuronal cell line (HT-22 cell) was used to assess the mechanism and mode of death produced by TMT. The compound produced a time- and concentration-dependent apoptotic death that was caspase-mediated. Excessive generation of reactive oxygen species (ROS) and subsequent reduction of mitochondrial membrane potential (DeltaPsim) were involved in the cytotoxicity. Scavenging of ROS by a free radical trapping agent or inhibition of the mitochondrial permeability transition (MPT) pore significantly reduced cell death. Additionally, TMT increased expression of inducible nitric oxide synthase (iNOS) by activation of the redox-sensitive transcription factor NFkappaB. Pharmacologic inhibition studies showed that the iNOS-mediated NO generation increased expression of Bax and then mitochondrial-mediated apoptosis. It was concluded that excessive ROS generation initiated the apoptotic cell death by upregulating iNOS followed by increased Bax expression which then led to loss of DeltaPsim and caspase-executed cell death. This study is the first to report in a neuronal cell model that TMT stimulates induction of iNOS, which then increases cellular levels of reactive nitrogen species (RNS) to initiate apoptotic death.
- Published
- 2006
- Full Text
- View/download PDF
15. PPARalpha-mediated upregulation of uncoupling protein-2 switches cyanide-induced apoptosis to necrosis in primary cortical cells.
- Author
-
Li L, Prabhakaran K, Zhang X, Borowitz JL, and Isom GE
- Subjects
- Animals, Base Sequence, DNA Primers, Necrosis, Pyrimidines pharmacology, RNA, Small Interfering, Rats, Rats, Sprague-Dawley, Reverse Transcriptase Polymerase Chain Reaction, Uncoupling Protein 2, Apoptosis drug effects, Cyanides pharmacology, Ion Channels physiology, Mitochondrial Proteins physiology, PPAR alpha physiology, Up-Regulation physiology
- Abstract
Peroxisome proliferator-activated receptor alpha (PPARalpha) is a member of the nuclear factor PPAR family that regulates a variety of cellular functions, including lipid metabolism, cellular oxidative stress defense, and inflammatory responses. Based on the report that Wy14,643, a PPARalpha agonist, can upregulate uncoupling protein-2 (UCP-2), this study was conducted in primary cortical cells to determine if PPARalpha activation enhances cyanide-induced neurotoxicity through changes in the level of UCP-2. PCR and Western blot analysis showed that Wy14,643 upregulated UCP-2 transcriptionally over a 12-h period. This response was mediated by PPARalpha since it was blocked by MK886, a selective PPARalpha antagonist. The effect of UCP-2 upregulation on the cytotoxic response to cyanide was quantitated by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (apoptosis) and propidium iodide staining (necrosis). Wy14,643 switched the mode of cyanide-induced cell death from apoptosis to necrosis. Cell death was preceded by marked mitochondrial dysfunction, as reflected by depletion of ATP and reduction of the mitochondrial membrane potential (DeltaPsim). Knock down of UCP-2 expression by RNA interference blocked the Wy14,643-mediated enhancement of cyanide-induced mitochondrial dysfunction and the switch of the cell death mode, thus confirming that the response was mediated by upregulation of UCP-2. This study shows that PPARalpha activation can upregulate UCP-2 expression, which in turn enhances cyanide-induced necrotic cell death through an increase of mitochondrial dysfunction.
- Published
- 2006
- Full Text
- View/download PDF
16. Up-regulation of uncoupling protein 2 by cyanide is linked with cytotoxicity in mesencephalic cells.
- Author
-
Prabhakaran K, Li L, Mills EM, Borowitz JL, and Isom GE
- Subjects
- Adenosine Triphosphate metabolism, Animals, Apoptosis drug effects, Cells, Cultured, Ion Channels, Mesencephalon pathology, Mitochondria drug effects, Mitochondria physiology, Necrosis, Rats, Rats, Sprague-Dawley, Uncoupling Protein 2, Up-Regulation, Cyanides toxicity, Membrane Transport Proteins physiology, Mesencephalon drug effects, Mitochondrial Proteins physiology
- Abstract
Uncoupling protein 2 (UCP-2) regulates mitochondrial function by increasing proton leak across the inner membrane to dissociate respiration from ATP synthesis and reduce reactive oxygen species generation. A number of studies have shown that UCP-2 expression protects cells from oxidative stress mediated injuries. In the current study, we show UCP-2-mediated reduction in mitochondrial function contributes to the mitochondrial dysfunction and the necrotic death of primary cultured mesencephalic cells (MCs) after exposure to cyanide, a complex IV inhibitor. The necrotic cell death was directly related to the level of mitochondrial dysfunction, as shown by reduction in ATP levels and decreased mitochondrial membrane potential. Treatment with cyanide for 6 h or longer upregulated UCP-2 expression. Blockade of up-regulation with a transcription or a translational inhibitor reduced the response to cyanide. Knockdown with RNAi or transfection with a UCP-2 dominant-negative interfering mutant reduced the cyanide-induced mitochondrial dysfunction and cell death, showing that constitutive expression of UCP-2 plays a role in the response to cyanide. Overexpression of UCP-2 by transfection with human full-length cDNA potentiated the cyanide toxicity. These findings indicate that UCP-2 can serve as a regulator of mitochondria-mediated necrotic cell death, in which enhanced expression can increase the vulnerability of primary MCs to injury due to complex IV-mediated inhibition by cyanide.
- Published
- 2005
- Full Text
- View/download PDF
17. Enhancement of cyanide-induced mitochondrial dysfunction and cortical cell necrosis by uncoupling protein-2.
- Author
-
Li L, Prabhakaran K, Mills EM, Borowitz JL, and Isom GE
- Subjects
- Animals, Apoptosis drug effects, Cells, Cultured, Cerebral Cortex cytology, Cerebral Cortex drug effects, Mitochondria physiology, Rats, Rats, Sprague-Dawley, Cyanides toxicity, Mitochondria drug effects
- 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.
- Published
- 2005
- Full Text
- View/download PDF
18. Receptor mechanisms mediating cyanide generation in PC12 cells and rat brain.
- Author
-
Gunasekar PG, Prabhakaran K, Li L, Zhang L, Isom GE, and Borowitz JL
- Subjects
- 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, Brain metabolism, Cyanides metabolism, Oxotremorine analogs & derivatives, PC12 Cells metabolism, Receptors, Cell Surface metabolism
- 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.
- Published
- 2004
- Full Text
- View/download PDF
19. Calcineurin-mediated Bad translocation regulates cyanide-induced neuronal apoptosis.
- Author
-
Shou Y, Li L, Prabhakaran K, Borowitz JL, and Isom GE
- Subjects
- Animals, Calcineurin Inhibitors, Calcium antagonists & inhibitors, Calcium metabolism, Calcium Signaling drug effects, Cells, Cultured, Chelating Agents metabolism, Chelating Agents pharmacology, Cyanides antagonists & inhibitors, Cytochromes c metabolism, Cytosol drug effects, Cytosol metabolism, Mitochondria drug effects, Mitochondria metabolism, Mitogen-Activated Protein Kinases metabolism, NF-kappa B metabolism, Neurons cytology, Neurons metabolism, Protein Transport drug effects, Rats, Rats, Sprague-Dawley, bcl-Associated Death Protein, p38 Mitogen-Activated Protein Kinases, Apoptosis drug effects, Calcineurin metabolism, Carrier Proteins metabolism, Cyanides pharmacology, Neurons drug effects
- Abstract
In cyanide-induced apoptosis, an increase in cytosolic free Ca2+ and generation of reactive oxygen species are initiation stimuli for apoptotic cell death. Previous studies have shown that cyanide-stimulated translocation of Bax (Bcl-associated X protein) to mitochondria is linked with release of cytochrome c and subsequent activation of a caspase cascade [Shou, Li, Prabhakaran, Borowitz and Isom (2003) Toxicol. Sci. 75, 99-107]. In the present study, the relationship of the cyanide-induced increase in cytosolic free Ca2+ to activation of Bad ( Bcl-2/Bcl-X(L)- antagonist, causing cell death) was determined in cortical cells. Bad is a Ca2+-sensitive pro-apoptotic Bcl-2 protein, which on activation translocates from cytosol to mitochondria to initiate cytochrome c release. In cultured primary cortical cells, cyanide produced a concentration- and time-dependent translocation of Bad from cytosol to mitochondria. Translocation occurred early in the apoptotic response, since mitochondrial Bad was detected within 1 h of cyanide treatment. Mitochondrial levels of the protein continued to increase up to 12 h post-cyanide exposure. Concurrent with Bad translocation, a Ca2+-sensitive increase in cellular calcineurin activity was observed. Increased cytosolic Ca2+ and calcineurin activation stimulated Bad translocation since BAPTA [bis-(o-aminophenoxy)ethane-N, N, N', N'-tetra-acetic acid], an intracellular Ca2+ chelator, and cyclosporin A, a calcineurin inhibitor, significantly reduced translocation. BAPTA also blocked release of cytochrome c from mitochondria as well as apoptosis. Furthermore, treatment of cells with the calcineurin inhibitors cyclosporin A or FK506 blocked the apoptotic response, linking calcineurin activation and the subsequent translocation of Bad to cell death. These observations show that by inducing a rapid increase in cytosolic free Ca2+, cyanide can partially initiate the apoptotic cascade through a calcineurin-mediated translocation of Bad to mitochondria.
- Published
- 2004
- Full Text
- View/download PDF
20. Caspase inhibition switches the mode of cell death induced by cyanide by enhancing reactive oxygen species generation and PARP-1 activation.
- Author
-
Prabhakaran K, Li L, Borowitz JL, and Isom GE
- Subjects
- Adenosine Triphosphate metabolism, Amino Acid Chloromethyl Ketones pharmacology, Animals, Caspase 3, Cell Death drug effects, Cells, Cultured, Cerebral Cortex cytology, Cerebral Cortex embryology, Cysteine Proteinase Inhibitors pharmacology, Enzyme Activation, Membrane Potentials drug effects, Mitochondria drug effects, Mitochondria physiology, Rats, Rats, Sprague-Dawley, Caspase Inhibitors, Poly(ADP-ribose) Polymerases metabolism, Potassium Cyanide toxicity, Reactive Oxygen Species metabolism
- Abstract
Execution of apoptosis can involve activation of the caspase family of proteases. Recent studies show that caspase inhibition can switch the morphology of cell death from apoptotic to necrotic without altering the level of death among cell populations. In the present study, the effect of caspase inhibition on cortical (CX) cell death induced by cyanide was investigated. In primary cultured CX cells exposed to cyanide (400 microM), death was primarily apoptotic as indicated by positive TUNEL staining. Reactive oxygen species (ROS) generation and subsequent caspase activation mediated the apoptosis. Inhibition of the caspase cascade with zVAD-fmk switched the apoptotic response to necrotic cell death, as assessed by increased cellular efflux of LDH and propidium iodide uptake by the cells. The change in death mode was accompanied by a marked increase in poly (ADP-ribose) polymerase-1 (PARP-1) activity, reactive oxygen species (ROS) generation, a reduction in the mitochondrial membrane potential (Delta psi(m)), and reduced cellular ATP. Prior treatment of cells with 3-aminobenzamide (3-AB), a PARP-1 inhibitor, prevented the cells from undergoing necrosis and preserved intracellular ATP levels. These findings indicate that apoptosis and necrosis share common initiation pathways and caspase inhibition can switch the apoptotic response to necrosis. Inhibition of PARP-1 preserves cellular ATP levels and in turn blocks execution of the necrotic death pathway.
- Published
- 2004
- Full Text
- View/download PDF
21. p38 Mitogen-activated protein kinase regulates Bax translocation in cyanide-induced apoptosis.
- Author
-
Shou Y, Li L, Prabhakaran K, Borowitz JL, and Isom GE
- Subjects
- 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, Apoptosis drug effects, Mitogen-Activated Protein Kinases metabolism, Potassium Cyanide toxicity, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-bcl-2
- 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.
- Published
- 2003
- Full Text
- View/download PDF
22. Cyanide enhancement of dopamine-induced apoptosis in mesencephalic cells involves mitochondrial dysfunction and oxidative stress.
- Author
-
Jones DC, Prabhakaran K, Li L, Gunasekar PG, Shou Y, Borowitz JL, and Isom GE
- Subjects
- Animals, Apoptosis physiology, Cells, Cultured, Membrane Potentials drug effects, Membrane Potentials physiology, Mesencephalon cytology, Mesencephalon metabolism, Mitochondria metabolism, Oxidative Stress physiology, Rats, Rats, Sprague-Dawley, Apoptosis drug effects, Cyanides pharmacology, Dopamine pharmacology, Mesencephalon drug effects, Mitochondria drug effects, Oxidative Stress drug effects
- Abstract
Dopamine (DA)-induced neurotoxicity is potentiated when cellular metabolism is compromised. Since cyanide is a neurotoxin that produces mitochondrial dysfunction and stimulates intracellular generation of reactive oxygen species (ROS), KCN was used to study DA-induced apoptosis in primary cultured mesencephalon cells. Treatment of neurons with DA (300 microM) for 24h produced apoptosis as determined by TUNEL staining, DNA fragmentation and increased caspase activity. Pretreatment with KCN (100 microM) 30min prior to DA increased the number of cells undergoing apoptosis. When added to the cells alone, this concentration of KCN did not induce apoptosis. DA stimulated intracellular generation of ROS, and treatment with KCN enhanced ROS generation. Treatment of cells with glutathione or uric acid (antioxidants/scavengers) attenuated both the increase in ROS generation and the apoptosis, demonstrating that ROS are initiators of the cytotoxicity. Studies on the sequence of events mediating the response showed that DA-induced depolarization of the mitochondrial membrane was dependent on ROS generation and KCN enhanced this action of DA. Following changes in mitochondrial membrane potential, cytochrome c was released from mitochondria, leading to caspase activation and eventually cell death. These results demonstrate that oxidative stress and mitochondrial dysfunction are initiators of DA-induced apoptosis. Subsequent cytochrome c release activates the caspase effector component of apoptosis. Cyanide potentiates the neurotoxicity of DA by enhancing the generation of ROS and impairing mitochondrial function.
- Published
- 2003
- Full Text
- View/download PDF
23. Oxidative stress and cyclooxygenase-2 induction mediate cyanide-induced apoptosis of cortical cells.
- Author
-
Li L, Prabhakaran K, Shou Y, Borowitz JL, and Isom GE
- Subjects
- Animals, Antioxidants pharmacology, Cerebral Cortex metabolism, Cerebral Cortex pathology, Cyclooxygenase 2, Cyclooxygenase 2 Inhibitors, Cyclooxygenase Inhibitors pharmacology, Dinoprostone biosynthesis, Enzyme Induction drug effects, NG-Nitroarginine Methyl Ester pharmacology, Rats, Rats, Sprague-Dawley, Apoptosis drug effects, Cerebral Cortex drug effects, Isoenzymes biosynthesis, Oxidative Stress, Potassium Cyanide toxicity, Prostaglandin-Endoperoxide Synthases biosynthesis
- Abstract
Cyanide (KCN)-induced generation of reactive oxygen species (ROS) involves cyclooxygenase-2 (COX-2)-mediated reactions in some neurons. The present study examines the extent to which COX isoforms are involved in KCN-induced apoptotic cell death processes of cultured cortical cells. After treatment with KCN (10-300 microM), COX-2 was expressed in a time- and concentration-dependent manner increasing markedly over a 4-h period. However, no significant changes were observed in COX-1 levels at any cyanide concentration. Correlated with COX-2 up-regulation, KCN induced a time-dependent apoptotic death. TUNEL staining showed that the COX-2 inhibitor NS-398 (30 microM) blocked KCN-induced apoptosis, whereas the selective COX-1 inhibitor valeryl salicylate did not affect the level of apoptotic cell death. Exposure of cells to KCN (300 microM) for 24 h resulted in DNA fragmentation, which was also reduced by NS-398. Prostaglandin E(2) (PGE(2)) accumulation in cell culture supernatants was increased by KCN and NS-398 blocked PGE(2) generation. PCR studies further confirmed that COX-2 expression was increased by KCN. Antioxidants phenyl-N-test-butylnitrone, superoxide dismutase, and catalase significantly inhibited KCN-induced COX-2 up-regulation and subsequent apoptosis. N(G)-nitro-L-arginine methylester an inhibitor of nitric oxide synthase, blocked KCN-induced PGE(2) production and apoptosis, but not COX-2 expression. Increased nitric oxide levels caused by cyanide may directly activate the COX-2 enzyme. These data show that cyanide treatment of cortical cells involves increased COX-2 expression, PGE(2) accumulation, and ROS generation, resulting in apoptotic cell death.
- Published
- 2002
- Full Text
- View/download PDF
24. Cyanide induces different modes of death in cortical and mesencephalon cells.
- Author
-
Prabhakaran K, Li L, Borowitz JL, and Isom GE
- Subjects
- Animals, Antioxidants pharmacology, Apoptosis drug effects, Blotting, Western, Caspase 3, Caspase Inhibitors, Caspases metabolism, Cell Death drug effects, Cells, Cultured, Cerebral Cortex drug effects, Cytochrome c Group metabolism, DNA biosynthesis, DNA isolation & purification, Electrophoresis, Polyacrylamide Gel, Enzyme Inhibitors pharmacology, In Situ Nick-End Labeling, Membrane Potentials drug effects, Mesencephalon drug effects, Mitochondria drug effects, Necrosis, Nitric Oxide Synthase metabolism, Nitric Oxide Synthase Type III, Rats, Cerebral Cortex pathology, Cyanides toxicity, Mesencephalon pathology
- Abstract
A comparative study was conducted in rat primary cortical (CX) and mesencephalic (MC) neurons to investigate intracellular cascades activated during cyanide-induced injury and to determine the point at which the cascades diverge to produce either apoptosis or necrosis. Cyanide treatment (400 microM) for 24 h produced primarily apoptosis in CX cells, whereas the same concentration of cyanide induced predominantly necrosis in MC cells as indicated by increased propidium iodide staining and cellular lactate dehydrogenase efflux. Cyanide increased generation of cellular reactive oxygen species (ROS) in both CX and MC cells, but the rate of formation and nature of the oxidative species varied with cell type. Catalase decreased cyanide-induced ROS generation in CX but not in MC cells. Nitric oxide generation was more prominent after cyanide treatment of MC compared with CX cells. N-Methyl-D-aspartate receptors were more involved in CX apoptosis than in MC necrosis. Mitochondrial membrane potential decreased moderately in CX cells on exposure to cyanide, whereas MC cells responded with a more pronounced reduction in potential. In CX cells cyanide produced a concentration-dependent release of cytochrome c from mitochondria and increased caspase activity, whereas little change was seen in MC neurons. Thus, cyanide-induced necrosis of MC cells involved generation of excessive amounts of nitric oxide and superoxide accompanied by mitochondrial depolarization. In contrast cyanide causes a lower level of oxidative stress in CX cells, involving mainly hydrogen peroxide and superoxide, and a moderate change in mitochondrial membrane potential that lead to cytochrome c release, caspase activation, and apoptosis.
- Published
- 2002
- Full Text
- View/download PDF
25. NF-kappaB-mediated up-regulation of Bcl-X(S) and Bax contributes to cytochrome c release in cyanide-induced apoptosis.
- Author
-
Shou Y, Li N, Li L, Borowitz JL, and Isom GE
- Subjects
- Animals, Blotting, Western, Caspases metabolism, DNA Fragmentation, Enzyme Inhibitors pharmacology, Mitochondria drug effects, Mitochondria metabolism, NF-kappa B antagonists & inhibitors, Neurons cytology, Neurons drug effects, Neurons metabolism, Oligonucleotides, Antisense pharmacology, Oxidation-Reduction, Peptides pharmacology, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-bcl-2 genetics, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Reverse Transcriptase Polymerase Chain Reaction, Up-Regulation drug effects, bcl-2-Associated X Protein, bcl-X Protein, Apoptosis drug effects, Cyanides toxicity, Cytochrome c Group metabolism, NF-kappa B metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism
- Abstract
Cyanide induces apoptosis through cytochrome c activated caspase cascade in primary cultured cortical neurons. The underlying mechanism for cytochrome c release from mitochondria after cyanide treatment is still unclear. In this study, the roles of endogenous Bcl-2 proteins in cyanide-induced apoptosis were investigated. After cyanide (100-500 microm) treatment for 24 h, two pro-apoptotic Bcl-2 proteins, Bcl-X(S) and Bax were up-regulated as shown by western blot and RT-PCR analysis. The expression levels of two antiapoptotic Bcl-2 proteins, Bcl-2 and Bcl-X(L), remained unchanged after cyanide treatment, whereas the mRNA levels of Bcl-X(S) and Bax began to increase within 2 h and their protein levels increased 6 h after treatment. NF-kappaB, a redox-sensitive transcription factor activated after cyanide treatment, is responsible for the up-regulation of Bcl-X(S) and Bax. SN50, which is a synthetic peptide that blocks translocation of NF-kappaB from cytosol to nucleus, inhibited the up-regulation of Bcl-X(S) and Bax. Similar results were obtained using a specific kappaB decoy DNA. NMDA receptor activation and reactive oxygen species (ROS) generation are upstream events of NF-kappaB activation, as blockade of these two events by MK801, l-NAME or PBN inhibited cyanide-induced up-regulation of Bcl-X(S) and Bax. Up-regulation of pro-apoptotic Bcl-X(S) and Bax contributed to cyanide-induced cytochrome c release, because SN50 and a specific Bax antisense oligodeoxynucleotide significantly reduced release of cytochrome c from mitochondria as shown by western blot analysis. It was concluded that NF-kappaB-mediated up-regulation of Bcl-X(S) and Bax is involved in regulating cytochrome c release in cyanide-induced apoptosis.
- Published
- 2002
- Full Text
- View/download PDF
26. Reactive oxygen species mediate pyridostigmine-induced neuronal apoptosis: involvement of muscarinic and NMDA receptors.
- Author
-
Li L, Shou Y, Borowitz JL, and Isom GE
- Subjects
- Animals, Animals, Newborn, Antioxidants pharmacology, Atropine pharmacology, Caspase 3, Caspases biosynthesis, Catalase pharmacology, Cells, Cultured, Cerebellum cytology, Cyclic N-Oxides, DNA drug effects, Dizocilpine Maleate pharmacology, Dose-Response Relationship, Drug, Electrophoresis, Agar Gel, In Situ Nick-End Labeling, L-Lactate Dehydrogenase metabolism, NG-Nitroarginine Methyl Ester pharmacology, Neurons enzymology, Neurons ultrastructure, Nitrogen Oxides pharmacology, Rats, Superoxide Dismutase pharmacology, Apoptosis drug effects, Cholinergic Agents metabolism, Neurons drug effects, Pyridostigmine Bromide toxicity, Reactive Oxygen Species metabolism, Receptors, N-Methyl-D-Aspartate metabolism
- Abstract
Pyridostigmine bromide (PB) is a reversible cholinesterase inhibitor used for treatment of myasthenia gravis and for prophylactic protection against organophosphate nerve agent. We previously showed PB can induce apoptotic death in rat brain following systemic treatment. To study mechanisms by which PB induces brain cell death, cultured rat cerebellar granule cells were used. Cytotoxicity was determined after exposure to PB (10-1000 microM) for 24 h; a high concentration of PB (>500 microM) significantly increased lactate dehydrogenase release, which was reduced by pretreatment with the antioxidant, N-t-butyl-alpha-phenyl-nitrone (PBN). Apoptosis, as determined by TUNEL staining, was concentration dependent (10-250 microM) after a 24-h exposure and cytotoxicity was confirmed by gel electrophoresis of DNA, release of cytochrome c from mitochondria, elevation of caspase activity, and electron microscopy. The oxidant-sensitive fluorescent dye 2',7'-dichlorofluorescin diacetate was used to detect reactive oxidative species (ROS) generation. Pretreatment with PBN, superoxide dismutase, catalase, or the nitric oxide synthase inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME) blocked PB-induced ROS generation and apoptotic cell death. Pretreatment with atropine or MK-801 blocked ROS generation and the subsequent neurotoxicity, showing that both muscarinic and NMDA receptors mediate the response. DNA extracted from PB-treated cells revealed oligonucleosomal fragmentation on gel electrophoresis and antioxidants attenuated the DNA fragmentation, providing further evidence for a link of ROS generation and apoptosis. These results indicate that muscarinic receptor-mediated ROS generation is an initiating factor in PB-induced apoptotic cell death and activation of the NMDA glutamate receptor is directly linked to the response., (Copyright 2001 Academic Press.)
- Published
- 2001
- Full Text
- View/download PDF
27. Mechanisms of the apoptotic and necrotic actions of trimethyltin in cerebellar granule cells.
- Author
-
Gunasekar P, Li L, Prabhakaran K, Eybl V, Borowitz JL, and Isom GE
- Subjects
- 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, Apoptosis drug effects, Cerebellum drug effects, Necrosis, Neurons drug effects, Trimethyltin Compounds toxicity
- 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.
- Published
- 2001
- Full Text
- View/download PDF
28. Role of astrocytes in trimethyltin neurotoxicity.
- Author
-
Gunasekar PG, Mickova V, Kotyzova D, Li L, Borowitz JL, Eybl V, and Isom GE
- Subjects
- Animals, Astrocytes metabolism, Astrocytes physiology, Cells, Cultured, Cerebellum cytology, Cerebellum drug effects, Cerebellum metabolism, Cytoplasmic Granules drug effects, Cytoplasmic Granules metabolism, Glutathione metabolism, Male, Mice, Oxidative Stress, Rats, Astrocytes drug effects, Trimethyltin Compounds toxicity
- Abstract
Although the neurotoxicity of trimethyltin (TMT) is well known, mechanisms are still not clear. Glia have been proposed to mediate the toxic action of TMT on nerve cells. Accordingly, the effects of TMT were tested in primary neuronal cultures from rat cerebellum and compared to effects in astrocytes and mixed cultures. Neuronal damage observed following TMT exposure was less in the presence of astrocytes and astrocytes alone were resistant to TMT. Thus, astrocytes have a protective effect against TMT-induced neurotoxicity. TMT caused an oxidative stress in granule cell cultures involving a variety of oxidative species (O2)*-, H2O2, NO), but astrocytes were less sensitive to TMT-induced oxidative species generation. Antioxidants, glutathione and 7-nitroindazole attenuated neuronal cell death induced by TMT. It appears that oxidative stress mediates a large part of the destructive action of TMT in neuronal cultures. The presence of astrocytes appears to modulate TMT-induced oxidative stress so that TMT causes only a small increase in lipid peroxidation in mouse brain after systemic administration. Thus, TMT induces a pronounced oxidative stress in cultured neurons, but when astrocytes are present, oxidative species play a lesser role in the neurotoxic action of TMT., (Copyright 2001 John Wiley & Sons, Inc. J Biochem Mol Toxicol 15:256–262, 2001)
- Published
- 2001
- Full Text
- View/download PDF
29. Endogenous generation of cyanide in neuronal tissue: involvement of a peroxidase system.
- Author
-
Gunasekar PG, Borowitz JL, Turek JJ, Van Horn DA, and Isom GE
- Subjects
- Aminobenzoates pharmacology, Analgesics, Opioid pharmacology, Animals, Azides pharmacology, Brain Chemistry drug effects, Carbachol pharmacology, Cell Fractionation, Cholinergic Agonists pharmacology, Dose-Response Relationship, Drug, Glycine pharmacology, Hydrogen Peroxide pharmacology, Hydromorphone pharmacology, Male, Mitochondria drug effects, Mitochondria metabolism, Morphine pharmacology, Narcotics pharmacology, Neurons cytology, Neurons drug effects, Oxidants pharmacology, PC12 Cells, Peroxidases antagonists & inhibitors, Rats, Rats, Sprague-Dawley, Cyanides metabolism, Neurons metabolism, Peroxidases metabolism
- Abstract
In a study of the mechanism by which cyanide is produced in neural tissue, it was hypothesized that nerve cells generate cyanide in a manner similar to that in leukocytes. As in white blood cells, glycine addition enhanced cyanide production in rat pheochromocytoma cells. Because myeloperoxidase catalyses cyanide production in leukocytes, a selective myeloperoxidase inhibitor (aminobenzoic acid hydrazide) was tested and found to inhibit opiate agonist-induced cyanide production in pheochromocytoma cells and also in rat brain. In addition, hydrogen peroxide enhanced cyanide release in pheochromocytoma cells, further suggesting that the process is oxidative in nature. Sonicated rat pheochromocytoma cells did not generate cyanide in response to an agonist acting on surface receptors even though disrupted cells responded to glycine. The mitochondrial fraction from rat brain produced more cyanide in response to glycine than any other fraction. Thus glycine seems to act at an intracellular site to enhance cyanide production and the process seems to involve a peroxidase mechanism similar to that reported for white blood cells., (Copyright 2000 Wiley-Liss, Inc.)
- Published
- 2000
- Full Text
- View/download PDF
30. Muscarinic receptor-mediated pyridostigmine-induced neuronal apoptosis.
- Author
-
Li L, Gunasekar PG, Borowitz JL, and Isom GE
- Subjects
- Animals, Brain drug effects, Brain enzymology, Cells, Cultured, Cerebral Cortex cytology, Cerebral Cortex drug effects, Cholinesterases metabolism, DNA Fragmentation drug effects, In Situ Nick-End Labeling, Male, Microscopy, Electron, Neurons ultrastructure, Rats, Rats, Sprague-Dawley, Apoptosis drug effects, Neurons drug effects, Parasympathomimetics pharmacology, Pyridostigmine Bromide pharmacology, Receptors, Muscarinic drug effects
- Abstract
Pyridostigmine is a reversible cholinesterase (ChE) inhibitor that is associated with neurologic dysfunction involving both central and peripheral nervous systems. To determine the neurotoxic potential of pyridostigmine, rats were sacrificed at intervals after drug administration (0.5-1.85 mg/kg, i.p., twice daily for 4 days) and brains examined histologically. ChE inhibition was used as a biomarker of pyridostigmine activity. Using the in situ terminal deoxynucleotidyl transferase nick-end labeling of DNA fragments (TUNEL) method and electron microscopy, apoptotic brain cell death was noted in cerebral cortex over a dose range of 0.5-1.85 mg/kg and at the higher dose (1.85 mg/kg), apoptosis was also noted in striatum and hippocampus. These responses were blocked by pretreatment with atropine. Rat cortical cells in culture also underwent apoptosis when exposed to pyridostigmine (250 microM for 24 hr), indicating that the pyridostigmine can initiate apoptosis, independent of peripheral mechanisms. Pretreatment of cells with atropine (10 microM) inhibited pyridostigmine-induced apoptosis, confirming the response was mediated by muscarinic receptors. Short term treatment of rats with pyridostigmine (1.85 mg/kg twice daily for 4 days) induced a prolonged apoptotic response, which was evident in rat cortex up to 30 days after the last dose. Active apoptosis persisted, despite recovery of serum ChE activity. These in vivo and in vitro observations indicate that pyridostigmine can initiate a prolonged neurodegeneration.
- Published
- 2000
31. Dopamine-induced apoptosis is mediated by oxidative stress and Is enhanced by cyanide in differentiated PC12 cells.
- Author
-
Jones DC, Gunasekar PG, Borowitz JL, and Isom GE
- Subjects
- Animals, Apoptosis physiology, Cell Differentiation, Cell Survival drug effects, Cell Survival physiology, Dose-Response Relationship, Drug, L-Lactate Dehydrogenase metabolism, Mitochondria drug effects, Mitochondria metabolism, Neurons drug effects, Neurons enzymology, Nitric Oxide metabolism, Oxidative Stress physiology, PC12 Cells, Rats, Apoptosis drug effects, Dopamine pharmacology, Enzyme Inhibitors pharmacology, Neurons cytology, Oxidative Stress drug effects, Potassium Cyanide pharmacology
- Abstract
Dopamine (DA) oxidation and the generation of reactive oxygen species (ROS) may contribute to the degeneration of dopaminergic neurons underlying various neurological conditions. The present study demonstrates that DA-induced cytotoxicity in differentiated PC12 cells is mediated by ROS and mitochondrial inhibition. Because cyanide induces parkinson-like symptoms and is an inhibitor of the antioxidant system and mitochondrial function, cells were treated with KCN to study DA toxicity in an impaired neuronal system. Differentiated PC12 cells were exposed to DA, KCN, or a combination of the two for 12-36 h. Lactate dehydrogenase (LDH) assays indicated that both DA (100-500 microM) and KCN (100-500 microM) induced a concentration- and time-dependent cell death and that their combination produced an increase in cytotoxicity. Apoptotic death, measured by Hoechst dye and TUNEL (terminal deoxynucleotidyltransferase dUTP nick end-labeling) staining, was also concentration- and time-dependent for DA and KCN. DA plus KCN produced an increase in apoptosis, indicating that KCN, and thus an impaired system, enhances DA-induced apoptosis. To study the mechanism(s) of DA toxicity, cells were pretreated with a series of compounds and incubated with DA (300 microM) and/or KCN (100 microM) for 24 h. Nomifensine, a DA reuptake inhibitor, rescued nearly 60-70% of the cells from DA- and DA plus KCN-induced apoptosis, suggesting that DA toxicity is in part mediated intracellularly. Pretreatment with antioxidants attenuated DA- and KCN-induced apoptosis, indicating the involvement of oxidative species. Furthermore, buthionine sulfoximine, an inhibitor of glutathione synthesis, increased the apoptotic response, which was reversed when cells were pretreated with antioxidants. DA and DA plus KCN produced a significant increase in intracellular oxidant generation, supporting the involvement of oxidative stress in DA-induced apoptosis. The nitric oxide synthase inhibitor N(G)-nitro-L-arginine methyl ester and the peroxynitrite scavenger uric acid blocked apoptosis and oxidant production, indicating involvement of nitric oxide. These results suggest that DA neurotoxicity is enhanced under the conditions induced by cyanide and involves both ROS and nitric oxide-mediated oxidative stress as an initiator of apoptosis.
- Published
- 2000
- Full Text
- View/download PDF
32. Cyanide-induced apoptosis involves oxidative-stress-activated NF-kappaB in cortical neurons.
- Author
-
Shou Y, Gunasekar PG, Borowitz JL, and Isom GE
- Subjects
- Animals, Caspases drug effects, Cerebral Cortex metabolism, DNA Fragmentation drug effects, Dizocilpine Maleate pharmacology, Drug Interactions, In Situ Nick-End Labeling, Neuroprotective Agents pharmacology, Rats, Rats, Sprague-Dawley, Apoptosis drug effects, Caspases metabolism, Cerebral Cortex drug effects, NF-kappa B drug effects, Oxidative Stress, Poisons toxicity, Potassium Cyanide toxicity, Reactive Oxygen Species metabolism
- Abstract
The central nervous system is one of the main target organs in cyanide toxicity. Primary cultured cortical neurons were used to study the cellular mechanisms underlying cyanide-induced cytotoxicity. After exposure to KCN (100-300 microM) for 24 h, cortical neurons underwent apoptosis as characterized by positive TUNEL staining. Reactive oxygen species (ROS) play an important role in cyanide-induced neuronal apoptosis; immediately after cyanide (100-300 microM) treatment, ROS generation was observed and continued to be elevated for up to 3 h. NMDA receptor activation and subsequent Ca(2+) influx contribute in part to cyanide-induced ROS formation, since the selective NMDA receptor antagonist MK801 and intracellular Ca(2+) chelator BAPTA blocked ROS generation. Interestingly, caspases, recently reported to be involved in neuronal apoptosis, play a role in the late phase of ROS production after cyanide stimulation. Z-VAD, a nonspecific caspase inhibitor, blocked ROS generated 1 h after cyanide treatment, but it had no effect on ROS generated immediately after cyanide treatment. Nuclear factor kappaB (NF-kappaB), a redox-sensitive transcription factor, was activated dose dependently after cyanide treatment. Blockade of ROS generation by MK801, Z-VAD, and various antioxidants also blocked the activation of NF-kappaB. SN50, a synthetic peptide which inhibits the nuclear translocation of NF-kappaB, blocked cyanide-induced apoptotic cell death. These results indicate that NF-kappaB plays an important role in cyanide-induced apoptosis in cortical neurons, and the caspases may contribute in part to the activation of NF-kappaB after cyanide treatment by inducing the late phase of ROS generation., (Copyright 2000 Academic Press.)
- Published
- 2000
- Full Text
- View/download PDF
33. Differential susceptibility of brain areas to cyanide involves different modes of cell death.
- Author
-
Mills EM, Gunasekar PG, Li L, Borowitz JL, and Isom GE
- Subjects
- Animals, Antioxidants pharmacology, Brain pathology, Cerebral Cortex drug effects, Male, Mice, Substantia Nigra drug effects, Apoptosis drug effects, Brain drug effects, Potassium Cyanide toxicity
- Abstract
We have demonstrated that cyanide (KCN) induces selective degeneration of dopaminergic neurons in mice and apoptotic cell death in cultured neurons. In the present study the mode of cyanide-induced cell death was determined in the susceptible brain areas. Mice were treated with KCN (6 mg/kg ip) or vehicle (saline) twice daily for 1 to 12 days. After 3 days of KCN treatment, two separate lesions were observed in coronal brain sections. Widespread DNA fragmentation in parietal and suprarhinal regions of the motor cortex was observed by the in situ terminal deoxynucleotide transferase nick-end labeling (TUNEL) technique. Pyknosis and chromatin condensation, morphological hallmarks of apoptotic cells, were observed in TUNEL-positive regions. On the other hand, in the substantia nigra (SN), KCN produced a progressive, bilateral necrotic lesion that was evident by 3 days of treatment. The SN lesion was circumscribed by a prominent ring of glial infiltration, as determined by glial-acidic fibrillary protein (GFAP) immunostaining. The extent of the SN lesion steadily increased with treatment duration, and DNA fragmentation was not observed over the 1- to 12-day period. On the other hand, cortical apoptosis was not associated with necrotic cell loss or astrogliosis. Pretreatment of animals with the antioxidant alpha-phenyl-tert-butyl nitrone (PBN) for 7 days prior to and during 3 days of KCN administration markedly reduced cortical DNA fragmentation whereas the PBN treatment did not influence the SN necrosis or astrocytic gliosis. Except for moderate GFAP immunostaining in corpus callosum, other brain areas were not affected by cyanide. It is concluded that KCN-induced neuronal loss involves selective activation of necrosis or apoptosis in different neuronal populations, and involves divergent mechanisms and sensitivity to antioxidants., (Copyright 1999 Academic Press.)
- Published
- 1999
- Full Text
- View/download PDF
34. Cyanide interaction with redox modulatory sites enhances NMDA receptor responses.
- Author
-
Sun P, Rane SG, Gunasekar PG, Borowitz JL, and Isom GE
- Subjects
- Animals, Calcium metabolism, Cells, Cultured, Cerebellum cytology, Cerebellum metabolism, N-Methylaspartate pharmacology, Neurons metabolism, Oxidation-Reduction, Protein Kinase C antagonists & inhibitors, Rats, Receptors, Glycine drug effects, Receptors, N-Methyl-D-Aspartate physiology, Cyanides toxicity, Neurons drug effects, Receptors, N-Methyl-D-Aspartate drug effects
- Abstract
Activation of NMDA receptors plays an important role in cyanide neurotoxicity. Cyanide indirectly activates the receptor by inducing neuronal release of glutamate and also enhances receptor-mediated responses by a direct interaction with the receptor complex. This study investigated the mechanism in cerebellar granule cells by which cyanide enhances NMDA-induced Ca2+ influx. Cyanide (50 microM) increased the influx of Ca2+ over the NMDA concentration range of 0.5-500 microM. Experiments showed that cyanide does not interact with the receptor's glycine or PKC mediated phosphorylation regulatory sites. N-ethylmaleimide, a thiol alkylating agent which inactivates the redox regulatory sites of the receptor, blocked the enhancing effect of cyanide. Pretreatment of cells with 5,5-dithio-bis-2-nitrobenzoic acid (DTNB), a compound that oxidizes the receptor redox sites, had no effect on the response to cyanide. On the other hand, the nonpermeant reducing agents, dithiothreitol or cysteine, further increased the cyanide effect. These observations can be explained by cyanide interacting with redox sensitive disulfide groups that are not accessible to the non-permeant reducing agents. It is proposed that cyanide interacts with a redox site(s) located either on the intracellular receptor domain or in the transmembrane hydrophobic domain. Furthermore the enhancement by cyanide of the excitotoxic actions of NMDA involves receptor sites that are sensitive to oxidation/reduction and this interaction contributes to the neurotoxic action of cyanide.
- Published
- 1999
- Full Text
- View/download PDF
35. Differential expression of group I metabotropic glutamate receptors (mGluRs) in the rat pheochromocytoma cell line PC12: role of nerve growth factor and ras.
- Author
-
Kane MD, Vanden Heuvel JP, Isom GE, and Schwarz RD
- Subjects
- Animals, Blotting, Western, Cell Differentiation physiology, Gene Expression drug effects, Gene Expression physiology, Neurons cytology, Neurons physiology, PC12 Cells, RNA, Messenger metabolism, Rats, Receptor, Metabotropic Glutamate 5, Receptors, Metabotropic Glutamate analysis, Reverse Transcriptase Polymerase Chain Reaction, Up-Regulation drug effects, Up-Regulation physiology, Nerve Growth Factors pharmacology, Neurons chemistry, Receptors, Metabotropic Glutamate genetics, ras Proteins physiology
- Abstract
Glutamate treatment of PC12 cells has been shown to result in the accumulation of intracellular inositol phosphates suggesting the presence of glutamate metabotropic receptors (mGluRs) positively coupled to phospholipase C. The present study examined the expression of group I mGluRs (mGluR1 and mGluR5) in PC12 cells. Undifferentiated PC12 cells were found to express both mGluR5 mRNA and receptor protein by reverse transcription polymerase chain reaction (RT-PCR) and western blot techniques. However, mGluR1 mRNA was not detected in these cells and western blot analysis showed only faint mGluR1alpha immunoreactivity suggesting a very low level of mGluR1 expression. Nerve growth factor-induced differentiation of PC12 cells resulted in the induction of mGluR1alpha and mGluR1beta mRNA and mGluR1alpha protein. PC12 cells overexpressing dominant negative ras revealed that NGF-induced mGluR1 induction, but not mGluR5 expression, is dependent on ras pathway activation in these cells. These results suggest PC12 cells may be a useful model for investigating the regulation and expression of group I mGluR isoforms and their role in neuronal processes in vitro.
- Published
- 1998
- Full Text
- View/download PDF
36. Cyanide-induced generation of oxidative species: involvement of nitric oxide synthase and cyclooxygenase-2.
- Author
-
Gunasekar PG, Borowitz JL, and Isom GE
- Subjects
- Animals, Cerebellar Cortex drug effects, Cerebellar Cortex enzymology, Cyclooxygenase 2, Isoenzymes metabolism, Nitric Oxide Synthase antagonists & inhibitors, Nitric Oxide Synthase metabolism, Phospholipases A antagonists & inhibitors, Phospholipases A metabolism, Phospholipases A2, Prostaglandin-Endoperoxide Synthases metabolism, Protein Kinase C antagonists & inhibitors, Protein Kinase C metabolism, Rats, Enzyme Inhibitors pharmacology, Isoenzymes drug effects, NG-Nitroarginine Methyl Ester pharmacology, Nitric Oxide Synthase drug effects, Potassium Cyanide pharmacology, Prostaglandin-Endoperoxide Synthases drug effects, Reactive Oxygen Species metabolism
- Abstract
In cerebellar granule cells, potassium cyanide (KCN) activates the NMDA receptor resulting in generation of nitric oxide and reactive oxygen species (ROS). To study the mechanism by which KCN stimulates ROS generation, the action of cyanide on the enzymatic pathways known to generate ROS were studied. The oxidant-sensitive fluorescent dye, 2,7-dichlorofluorescin was used to measure intracellular levels of nitric oxide and ROS in cerebellar granule cells. Using selective enzyme inhibitors, it was shown that both protein kinase C and phospholipase A2 are involved in KCN-stimulated generation of NO and ROS. In cells treated with indomethacin or nordihydroguairetic acid, inhibitors of cyclooxygenase (COX) and lipoxygenase (LOX) respectively, attenuated (approximately 35%) KCN-induced generation of oxidant species. When L-NAME (LG-nitro-L-arginine methyl ester) (nitric oxide synthase inhibitor, NOS) was combined with either indomethacin or nordihydroguairetic acid, generation of oxidant species was blocked by more than 80%. Pretreatment with NS398 (COX-2 inhibitor) significantly decreased ROS generation indicating the involvement of COX-2 in KCN-induced oxidant generation. Treatment with L-NAME + NS398 blocked oxidant species generation, reflecting involvement of NOS. The participation of cytochrome P450 was not evident because SKF525A did not significantly reduce KCN-induced ROS generation. Furthermore, a correlation was observed between oxidant generation and lipid peroxidation of cellular membranes (as determined by thiobarbituric acid levels). Pretreatment with inhibitors of protein kinase C, phospholipase A2 or COX, LOX, COX-2 partially blocked KCN-induced formation of thiobarbituric acid reactive substance, whereas coincubation of L-NAME with the inhibitors decreased lipid peroxidation by 60 to 90%. In cytotoxicity studies, KCN-induced cell death was partially blocked by the inhibitors and significant protection was observed when L-NAME was combined with these compounds. These findings show that activation of phospholipase A2 and subsequent metabolism of arachidonic acid by the COX-2 and LOX pathways and NOS contribute to cyanide-induced ROS production.
- Published
- 1998
37. Trimethyltin stimulates protein kinase C translocation through receptor-mediated phospholipase C activation in PC12 cells.
- Author
-
Kane MD, Yang CW, Gunasekar PG, and Isom GE
- Subjects
- Animals, Cell Differentiation, Enzyme Activation, Estrenes pharmacology, Excitatory Amino Acid Antagonists pharmacology, Glycine pharmacology, Inositol 1,4,5-Trisphosphate metabolism, Kinetics, Muscarinic Antagonists pharmacology, PC12 Cells, Protein Kinase C antagonists & inhibitors, Pyrrolidinones pharmacology, Rats, Receptors, Metabotropic Glutamate antagonists & inhibitors, Atropine pharmacology, Benzoates pharmacology, Glycine analogs & derivatives, Protein Kinase C metabolism, Trimethyltin Compounds pharmacology
- Abstract
Trimethyltin (TMT) is a potent neurotoxic compound that initiates a delayed neuronal cell death. Previously we have shown that TMT-induced cytotoxicity is associated with protein kinase C (PKC) translocation and activation. The present study investigates the mechanism underlying TMT-stimulated PKC translocation in PC12 cells. TMT exposure led to a rapid increase in intracellular levels of inositol 1,4,5-trisphosphate (IP3), a product of phospholipase C (PLC). This was significantly decreased by pretreating cells with antagonists to either the cholinergic muscarinic receptor (atropine) or the glutamatergic metabotropic receptor [(+)-alpha-methyl-4-carboxyphenylglycine; (+)-MCPG]. Furthermore, the rise in IP3 level was blocked by pretreating cells with a PLC inhibitor (U-73122) or by a combination of atropine and (+)-MCPG. This pretreatment also significantly decreased TMT-stimulated PKC translocation, indicating that TMT-mediated PKC translocation was related to PLC activation, presumably through formation of diacylglycerol, an endogenous activator of PKC and product of PLC. It is interesting that atropine and (+)-MCPG did not provide protection against TMT-induced cytotoxicity in these cells. However, these data suggest that TMT causes the release of cellular constituents that activate G protein-coupled receptors, ultimately leading to PKC translocation.
- Published
- 1998
- Full Text
- View/download PDF
38. Reactive oxygen species generated by cyanide mediate toxicity in rat pheochromocytoma cells.
- Author
-
Kanthasamy AG, Ardelt B, Malave A, Mills EM, Powley TL, Borowitz JL, and Isom GE
- Subjects
- Amitrole pharmacology, Animals, Ascorbic Acid pharmacology, Catalase antagonists & inhibitors, Cell Death drug effects, Dibucaine pharmacology, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, Lipid Peroxidation drug effects, Microscopy, Confocal, PC12 Cells metabolism, PC12 Cells pathology, Quinacrine pharmacology, Rats, Superoxide Dismutase antagonists & inhibitors, PC12 Cells drug effects, Potassium Cyanide toxicity, Reactive Oxygen Species
- Abstract
Peroxide formation has been implicated in impairment of motor function by cyanide which occurs in both animals and man. The present study employs the neuronal model, rat pheochromocytoma (PC12) cells to evaluate peroxidation as a toxic mechanism of cyanide. Confocal imaging shows that peroxides form within a few seconds in cell cytoplasm after cyanide exposure and continue to accumulate over a period of several minutes. Peroxide generation by cyanide is decreased to about 50% by phospholipase A2 inhibitors indicating involvement of arachidonic acid in the oxidative process. Also antioxidant defense enzymes (CuZn superoxide dismutase and especially catalase) in PC12 cells are inhibited by cyanide. It appears that peroxide accumulation after cyanide treatment involves both inhibition of breakdown and increased production. Furthermore, both peroxide accumulation and cell death induced by cyanide in PC12 cells are blocked by an antioxidant (ascorbate). These data support the hypothesis that the cytotoxic action of cyanide is related in part to an oxidative process.
- Published
- 1997
- Full Text
- View/download PDF
39. Hydrogen cyanide generation by mu-opiate receptor activation: possible neuromodulatory role of endogenous cyanide.
- Author
-
Borowitz JL, Gunasekar PG, and Isom GE
- Subjects
- 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, Hydrogen Cyanide pharmacology, Neurotransmitter Agents pharmacology, Receptors, Opioid, mu agonists
- 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.
- Published
- 1997
- Full Text
- View/download PDF
40. Modulation of the NMDA receptor by cyanide: enhancement of receptor-mediated responses.
- Author
-
Sun P, Rane SG, Gunasekar PG, Borowitz JL, and Isom GE
- Subjects
- 2-Amino-5-phosphonovalerate pharmacology, Animals, Calcium metabolism, Cells, Cultured, Cerebellum cytology, Cerebellum drug effects, Cerebellum metabolism, Cytoplasmic Granules drug effects, Cytoplasmic Granules metabolism, Diltiazem pharmacology, Dizocilpine Maleate pharmacology, Magnesium pharmacology, Membrane Potentials drug effects, N-Methylaspartate pharmacology, Nifedipine pharmacology, Patch-Clamp Techniques, Potassium metabolism, Rats, Receptors, N-Methyl-D-Aspartate metabolism, Tetrodotoxin pharmacology, Cyanides pharmacology, Receptors, N-Methyl-D-Aspartate drug effects
- Abstract
The effect of cyanide on the N-methyl-D-aspartate (NMDA)-stimulated increase in cytosolic free calcium ([Ca++]i) was studied by microfluorescence in fura-2-loaded cerebellar granule cells. The response to NMDA was enhanced by NaCN over a concentration range of 20 to 100 microM. These concentrations of NaCN in the absence of NMDA had no effect on basal [Ca++]i. In comparison, NaCN did not affect K+-depolarization-induced [Ca++]i elevation. The NaCN potentiation of NMDA-evoked [Ca++]i elevation was blocked by addition of Mg++ and by the NMDA receptor antagonists 2-amino-5-phosphono-valeric acid and (+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohept-5,10-imine maleate. Pretreatment of the cells with pregnenolone sulfate or arachidonate, known modulators of the NMDA receptor, enhanced NaCN action. The voltage-sensitive calcium channel blockers nifedepine and diltiazem did not affect the NaCN-induced potentiation. Additionally, the NaCN action was not altered when tetrodotoxin was used to block Na+ channel-mediated glutamate release. In patch-clamp studies, NaCN increased the amplitude and duration of NMDA-stimulated whole-cell currents. NaCN also enhanced the NMDA receptor response in single-channel patch-clamp experiments. In the outside-out patch recording configuration, NaCN increased the NMDA receptor channel opening frequency without affecting single-channel conductance or mean channel open time. These results indicate that cyanide interacts directly with the NMDA receptor channel complex to enhance receptor-mediated responses.
- Published
- 1997
41. 2-Iminothiazolidine-4-carboxylic acid produces hippocampal CA1 lesions independent of seizure excitation and glutamate receptor activation.
- Author
-
Bitner RS, Yim GK, and Isom GE
- Subjects
- 6-Cyano-7-nitroquinoxaline-2,3-dione therapeutic use, Animals, Brain Ischemia chemically induced, Dizocilpine Maleate therapeutic use, Glutamic Acid toxicity, Hippocampus blood supply, Injections, Intraventricular, Male, Proline toxicity, Rats, Rats, Sprague-Dawley, Brain Ischemia physiopathology, Excitatory Amino Acid Agonists therapeutic use, Hippocampus drug effects, Neuroprotective Agents therapeutic use, Neurotoxins toxicity, Seizures chemically induced
- Abstract
We previously demonstrated that 2-iminothiazolidine-4-carboxylic acid (2-ICA), formed by cyanide reacting with cysteine, caused glutamate antagonist-sensitive seizures when injected i.c.v. (intracerebroventricular) in mice and produced hippocampal CA1 damage following i.c.v. infusion in rats. In this study, the ability of either 2-ICA, glutamate, proline or NMDA (N-methyl-D-aspartate) injected i.c.v. to produce hippocampal lesions sensitive to glutamate antagonists was compared in mice. Hippocampal CA1 damage was observed 5-days following either a seizure (3.2 mumol) or subseizure (1.0 mumol) dose of 2-ICA. Glutamate (3.2 mumol) or proline (10 mumol) also produced hippocampal damage; glutamate damage was primarily to the CA1 subfield, whereas proline damaged neurons throughout the entire hippocampal formation. NMDA (3.2 nmol) caused seizure activity in all animals with a 50% lethality. No hippocampal damage was observed in surviving mice. Neither MK-801 (dizocilpine maleate) nor CNQX (6-cyano-7-nitroquinoxaline-2,3-dione) pretreatment prevented hippocampal lesions produced by 2-ICA. In contrast, MK-801 significantly reduced the frequency of mice displaying glutamate hippocampal lesions, but failed to block seizures produced by glutamate. MK-801 also protected neurons in the CA2-3 zone and the dentate gyrus, but not in the CA1 region of proline-injected mice. Finally, pretreatment with the mixed metabotropic glutamate receptor (mGluR)1/mGluR2 antagonist-agonist (S)-4-carboxy-3-hydroxyphenylglycine (CHPG) prevented hippocampal damage produced by the mGluR1 agonist (RS)-3,5-dihydroxyphenylglycine (DHPG), but did not protect against 2-ICA hippocampal lesions. These results show that 2-ICA hippocampal CA1 damage is not mediated through ionotropic or metabotropic glutamate receptors. 2-ICA hippocampal damage may represent a neurotoxicity that is distinct from excitotoxic-mediated cell death.
- Published
- 1997
42. Cyanide-induced apoptosis and oxidative stress in differentiated PC12 cells.
- Author
-
Mills EM, Gunasekar PG, Pavlakovic G, and Isom GE
- Subjects
- Animals, Ascorbic Acid pharmacology, Biotin, Blood Proteins pharmacology, Catalase pharmacology, Cell Differentiation drug effects, Cell Differentiation physiology, DNA Damage, Deoxyuracil Nucleotides, Endonucleases antagonists & inhibitors, Enzyme Inhibitors pharmacology, Fluorescent Dyes, Microscopy, Electron, Nucleosomes drug effects, PC12 Cells cytology, PC12 Cells ultrastructure, Rats, Reactive Oxygen Species metabolism, Staining and Labeling, Apoptosis drug effects, Oxidative Stress drug effects, PC12 Cells drug effects, Potassium Cyanide toxicity
- Abstract
Terminally differentiated PC12 cells are a useful neuron-like model for studying programmed cell death in response to nerve growth factor (NGF) deprivation. This in vitro model was used to investigate the mechanism by which cyanide-induced histotoxic hypoxia produces neuronal degeneration. Treatment of undifferentiated PC12 cells with 0.1 mM KCN for 24 h did not produce cell death. In contrast, treatment of differentiated PC12 cell cultures with 0.1 mM KCN for 24 h increased cell death by 43% when compared with control cultures, as measured by trypan blue dye exclusion and lactate dehydrogenase release assays. The Ca2+/Mg(2+)-dependent endonuclease inhibitor aurintricarboxylic acid and the transcriptional inhibitor actinomycin D partially attenuated hypoxic toxicity, suggesting roles for endonuclease activation and transcription in this model of neuronal death. Extracted DNA from cyanide-treated neurons demonstrated cleavage into oligonucleosomal fragments on gel electrophoresis. Transmission electron microscopic analysis showed morphological changes consistent with apoptotic cell death, including membrane blebbing and convolution, as well as chromatin condensation and margination to the nuclear membrane. Addition of either ascorbate or catalase to the cultures partially attenuated the loss of cell viability induced by cyanide, and decreased the incidence of apoptotic cells after treatment, based on the in situ detection of DNA strand breaks. The ability of cyanide to elevate intracellular oxidant species was determined by microfluorescence in differentiated PC12 cells loaded with the oxidant-sensitive dye 2',7'-dichlorofluorescin. Exposure of cells to 0.1 mM KCN produced a rapid generation of oxidants that was blocked approximately 50% by ascorbate or catalase. These observations indicate that cyanide induces apoptosis in terminally differentiated, and not undifferentiated, PC12 cells, and that antioxidants significantly reduce the incidence of cyanide-induced apoptosis.
- Published
- 1996
- Full Text
- View/download PDF
43. Cyanide-induced neurotoxicity involves nitric oxide and reactive oxygen species generation after N-methyl-D-aspartate receptor activation.
- Author
-
Gunasekar PG, Sun PW, Kanthasamy AG, Borowitz JL, and Isom GE
- Subjects
- Animals, Arginine analogs & derivatives, Arginine pharmacology, Calcium physiology, Dizocilpine Maleate pharmacology, Free Radicals, Lipid Peroxidation drug effects, NG-Nitroarginine Methyl Ester, Rats, Cerebellum drug effects, Nitric Oxide physiology, Potassium Cyanide toxicity, Reactive Oxygen Species metabolism, Receptors, N-Methyl-D-Aspartate drug effects
- Abstract
To study oxidative mechanisms in cyanide toxicity, cyanide-induced generation of intracellular oxidant species was determined by microfluorescence in cerebellar granule cells loaded with the oxidant-sensitive fluorescence dye 2,7-dichlorofluorescin. KCN produced a concentration-dependent (25-200 microM) generation of intracellular oxidant species that was blocked by N-methyl-D-aspartate receptor antagonists (MK-801 or AP5) or by removal of extracellular Ca++ from the incubation medium. To determine the relative contribution of NO and reactive oxygen species (ROS) to the increase of cellular fluorescence after KCN, a selective inhibitor of nitric oxide synthase, a NO scavenger and enzymes that metabolize ROS were added to the incubation medium. Interference with the nitric oxide system (reduced hemoglobin as a NO scavenger or [N(G)-nitro-L-arginine methyl ester [L-NAME] reduced fluorescence by 50%). Addition of enzymes that metabolize peroxide (catalase or superoxide dismutase [SOD]) also reduced fluorescence by nearly 50%. Combination of SOD with hemoglobin or L-NAME provided additional attenuation of the fluorescence and it was concluded that both NO and ROS are generated concurrently after KCN. Furthermore a correlation was observed between NO and ROS formation and levels of malonaldehyde (MDA), a marker of lipid peroxidation. Pretreatment with MK-801 blocked KCN-induced MDA formation, whereas L-NAME partially diminished MDA production. Treatment with a combination of SOD/catalase and L-NAME blocked the KCN-induced lipid peroxidation. In cytotoxicity studies cyanide-induced cell death was blocked by MK-801, whereas partial attenuation was produced by L-NAME; SOD/catalase treatments did not protect the cells. However, significant protection from cyanide-induced cytotoxicity was observed when L-NAME was combined with SOD/catalase. It is concluded that cyanide activates N-methyl-D-aspartate receptors to simultaneously generate both NO and ROS, which may lead to formation of the cytotoxic peroxynitrite anion.
- Published
- 1996
44. Cyanide-stimulated inositol 1,4,5-trisphosphate formation: an intracellular neurotoxic signaling cascade.
- Author
-
Yang CW, Borowitz JL, Gunasekar PG, and Isom GE
- Subjects
- Animals, Calcium metabolism, Cytosol metabolism, Egtazic Acid analogs & derivatives, Egtazic Acid pharmacology, PC12 Cells metabolism, Rats, Type C Phospholipases antagonists & inhibitors, Type C Phospholipases metabolism, Inositol 1,4,5-Trisphosphate biosynthesis, PC12 Cells drug effects, Poisons toxicity, Potassium Cyanide toxicity, Signal Transduction drug effects
- Abstract
Cyanide-induced neurotoxicity is associated with altered cellular Ca2+ homeostasis resulting in sustained elevation of cytosolic Ca2+. In order to characterize the effect of cyanide on intracellular signaling mechanisms, the interaction of KCN with the inositol 1,4,5-trisphosphate Ca2+ signaling system was determined in the PC12 cell line. KCN in the concentration range of 1.0-100 microM produced a rapid rise in intracellular IP3 levels (peak level occurred within 60 sec); 10 microM KCN elevated intracellular levels of IP3 to 148% of control levels. This response was mediated by phospholipase C (PLC) since U73122, a specific PLC inhibitor, blocked the response. Removal of Ca2+ from the incubation medium and chelation of intracellular Ca2+ with BAPTA partially attenuate the cyanide-stimulated IP3 generation, showing that the response is partially Ca2+ dependent. Also, treatment of cells with nifedipine or LaCl3, Ca2+ channel blockers, partially blocked the generation of IP3. This study shows that cyanide in concentrations as low as 1 microM stimulates IP3 generation that may be mediated by receptor and nonreceptor IP3 production since they have differential dependence on Ca2+. It is proposed that this response is an early intracellular signaling action that can contribute to altered Ca2+ homeostasis characteristic of cyanide neurotoxicity.
- Published
- 1996
- Full Text
- View/download PDF
45. Antagonism of cyanide toxicity by isosorbide dinitrate: possible role of nitric oxide.
- Author
-
Sun P, Borowitz JL, Kanthasamy AG, Kane MD, Gunasekar PG, and Isom GE
- Subjects
- Administration, Oral, Analysis of Variance, Animals, Injections, Intraperitoneal, Lethal Dose 50, Male, Methemoglobin metabolism, Methylene Blue chemistry, Methylene Blue metabolism, Mice, Mitochondria drug effects, Nitric Oxide biosynthesis, Oxygen Consumption drug effects, Potassium Cyanide administration & dosage, Potassium Cyanide toxicity, Sodium Nitrite administration & dosage, Sodium Nitrite pharmacology, Antidotes pharmacology, Isosorbide Dinitrate pharmacology, Nitric Oxide physiology, Poisons administration & dosage, Poisons toxicity, Potassium Cyanide antagonists & inhibitors, Respiration drug effects, Vasodilator Agents pharmacology
- Abstract
In a search for improved cyanide antidotes, the efficacy of isosorbide dinitrate (ISDN), was compared with that of the known cyanide antidote, NaNO2. ISDN was as effective as an optimal dose of NaNO2 in protecting mice against cyanide lethality. To study the mechanism involved, the extent of formation of the cyanide scavenger, methemoglobin, in the action of ISDN was determined. ISDN (300 mg/kg, p.o.) increased methemoglobin from 5 to 10% of total hemoglobin, while, in contrast, NaNO2 (100 mg/kg, i.p.) increased methemoglobin levels to 50% of total hemoglobin. Lowering the dose of NaNO2 to 30 mg/kg reduced methemoglobin levels to approximately 10% of total hemoglobin and in turn nearly abolished its antidotal effect. Decreasing methemoglobin to less than control levels using methylene blue failed to abolish cyanide antagonism by ISDN. Thus, methemoglobin formation by ISDN does not account for its antidotal action. Further studies comparing the respiratory depressant effects of cyanide in the presence of ISDN or NaNO2 also indicated that these two antidotes have different mechanisms of action. Efforts to produce tolerance to the antidotal effect of ISDN against cyanide toxicity were unsuccessful. It is suggested that the well-known ability of ISDN to generate nitric oxide may account for the noted cyanide antagonism.
- Published
- 1995
- Full Text
- View/download PDF
46. Modification of cyanide toxicodynamics: mechanistic based antidote development.
- Author
-
Isom GE and Borowitz JL
- Subjects
- Animals, Humans, Isosorbide Dinitrate pharmacology, Nitric Oxide physiology, Reactive Oxygen Species toxicity, Antidotes pharmacology, Cyanides toxicity
- Abstract
Cyanide's actions are complex and cannot be attributed solely to inhibition of oxygen utilization. Recent mechanistic studies show that cyanide inhibits multiple enzymes and alters several vital intracellular processes to produce the intoxication syndrome. By understanding the intracellular targets and the mechanisms underlying the toxicity, it is proposed that more effective antidotal regimens can be achieved. A mechanistic based, multi-step in vitro model was developed for screening potential cyanide antidotes. A series of compounds was screened for their ability to reverse the effect of cyanide on six neurochemical markers in the PC12 cell line (neuronal cell model). Each compound was assigned a composite score based on the six assays; several compounds were identified which then exhibited efficacy in animal testing. Additional mechanistic based studies show that antioxidants and nitric oxide generators have promise as anti-cyanide agents. It is concluded that mechanistic based antidote design can be used to identify new compounds for testing in animal models.
- Published
- 1995
- Full Text
- View/download PDF
47. Role of intracellular Cd2+ in catecholamine release and lethality in PC12 cells.
- Author
-
Kanthasamy AG, Isom GE, and Borowitz JL
- Subjects
- 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester pharmacology, Animals, Calcium Channel Agonists pharmacology, Calcium Channel Blockers pharmacology, Calcium Channels drug effects, Calcium Channels metabolism, Humans, Kinetics, Neurons cytology, Neurons metabolism, PC12 Cells, Rats, Cadmium metabolism, Cadmium toxicity, Cell Death drug effects, Dopamine metabolism, Neurons drug effects
- Abstract
To evaluate the role of intracellular Cd2+ in catecholamine release and lethality in rat pheochromocytoma (PC12) cells the following results were obtained: [1] the presence of Cd2+ intracellularly was demonstrated with the Cd(2+)-sensitive fluorescent dye BTC-5N, [2] Cd2+ entry through Ca(2+)-channels was either blocked with nifedipine or diltiazem or increased with Bay K8644, [3] Cd2+ entry through voltage sensitive Ca2+ channels was related to dopamine release and cell lethality, [4] a calmodulin inhibitor protected against Cd2+ toxicity, and [5] extracellular Ca2+ concentration, altered prior to Cd2+ exposure, was inversely related to dopamine release by Cd2+. The data indicate intracellular effects of Cd2+ rather than cell surface actions are primarily involved in neurotransmitter release and lethality by toxic levels of Cd2+ in adrenomedullary cells. To evaluate the role of intracellular Cd2+ in catecholamine release and lethality in rat pheochromocytoma (PC12) cells the following results were obtained: [1] the presence of Cd2+ intracellularly was demonstrated with the Cd(2+)-sensitive fluorescent dye BTC-5N, [2] Cd2+ entry through Ca(2+)-channels was either blocked with nifedipine or diltiazem or increased with Bay K8644, [3] Cd2+ entry through voltage sensitive Ca2+ channels was related to dopamine release and cell lethality, [4] a calmodulin inhibitor protected against Cd2+ toxicity, and [5] extracellular Ca2+ concentration, altered prior to Cd2+ exposure, was inversely related to dopamine release by Cd2+. The data indicate intracellular effects of Cd2+ rather than cell surface actions are primarily involved in neurotransmitter release and lethality by toxic levels of Cd2+ in adrenomedullary cells.
- Published
- 1995
- Full Text
- View/download PDF
48. NMDA receptor activation produces concurrent generation of nitric oxide and reactive oxygen species: implication for cell death.
- Author
-
Gunasekar PG, Kanthasamy AG, Borowitz JL, and Isom GE
- Subjects
- Animals, Calcium physiology, Cell Death physiology, Cells, Cultured, Cerebellum cytology, Cerebellum drug effects, Cerebellum metabolism, Extracellular Space metabolism, Fluoresceins metabolism, Glutamic Acid pharmacology, L-Lactate Dehydrogenase metabolism, Neuroprotective Agents pharmacology, Oxidation-Reduction, Protein Kinase C physiology, Quinacrine pharmacology, Rats, Nitric Oxide biosynthesis, Reactive Oxygen Species metabolism, Receptors, N-Methyl-D-Aspartate metabolism
- Abstract
The ability of glutamate to stimulate generation of intracellular oxidant species was determined by microfluorescence in cerebellar granule cells loaded with the oxidant-sensitive fluorescent dye 2,7-dichlorofluorescin (DCF). Exposure of cells to glutamate (10 microM) produced a rapid generation of oxidants that was blocked approximately 70% by MK-801 (a noncompetitive NMDA-receptor antagonist). To determine if nitric oxide (NO) or reactive oxygen species (ROS) contributed to the oxidation of DCF, cells were treated with compounds that altered their generation. NO production was inhibited with NG-nitro-L-arginine methyl ester (L-NAME) (nitric oxide synthase inhibitor) and reduced hemoglobin (NO scavenger). Alternatively, cells were incubated with superoxide dismutase (SOD) and catalase, which selectively metabolize O2-. and H2O2. Concurrent inhibition of O2-. and NO production nearly abolished intracellular oxidant generation. Pretreatment of cells with either chelerythrine (1 microM, protein kinase C inhibitor) or quinacrine (5 microM, phospholipase A2 inhibitor) before addition of glutamate also blocked oxidation of DCF. Generation of oxidants by glutamate was significantly reduced by incubating the cells of Ca(2+)-free buffer. In cytotoxicity studies, a positive correlation was observed between glutamate-induced death and oxidant generation. Glutamate-induced cytotoxicity was blocked by MK-801 and attenuated by treatment with L-NAME, chelerythrine, SOD, or quinacrine. It is concluded that glutamate induces concurrent generation of NO and ROS by activation of both NMDA receptors and non-NMDA receptors through a Ca(2+)-mediated process. Activation of NO synthase and phospholipase A2 contribute significantly to this response. It is proposed that simultaneous generation of NO and ROS results in formation of peroxynitrite, which initiates the cellular damage.
- Published
- 1995
- Full Text
- View/download PDF
49. Activation of protein kinase C by trimethyltin: relevance to neurotoxicity.
- Author
-
Pavlaković G, Kane MD, Eyer CL, Kanthasamy A, and Isom GE
- Subjects
- Alkaloids, Animals, Benzophenanthridines, Blotting, Western, Cell Death, Cell Differentiation, Enzyme Activation, Immunohistochemistry, Microscopy, Confocal, PC12 Cells drug effects, PC12 Cells metabolism, Phenanthridines pharmacology, Protein Kinase C antagonists & inhibitors, Rats, Tissue Distribution, Neurotoxins pharmacology, Protein Kinase C metabolism, Trimethyltin Compounds pharmacology
- Abstract
The differentiated PC12 cell neuronal model was used to determine the effect of trimethyltin (TMT) on protein kinase C (PKC). Cells treated with 5-20 microM TMT showed a partial and sustained PKC translocation within 30 min and persisted over a 24-h period. TMT treatment was accompanied by a low level of PKC down-regulation over 24 h, which was small compared with that produced by phorbol esters. Confocal imaging of differentiated PC12 cells showed that PKC translocates to the plasma membrane and the translocation is blocked by the PKC inhibitor chelerythrine (1 microM). Phorbol myristate-induced PKC down-regulation or inhibition with chelerythrine provided protection against TMT-induced cytotoxicity. It was concluded that TMT-induced PKC translocation and activation contribute to the cytotoxicity of TMT in differentiated PC12 cells.
- Published
- 1995
- Full Text
- View/download PDF
50. Monitoring intracellular nitric oxide formation by dichlorofluorescin in neuronal cells.
- Author
-
Gunasekar PG, Kanthasamy AG, Borowitz JL, and Isom GE
- Subjects
- Analysis of Variance, Animals, Cells, Cultured metabolism, Neurons metabolism, Nitric Oxide metabolism, Rats, Fluoresceins, Nitric Oxide biosynthesis, PC12 Cells metabolism
- 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.
- Published
- 1995
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.