Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Mol Biol Rep ; 48(5): 4517-4525, 2021 May.
Article in English | MEDLINE | ID: mdl-34089465

ABSTRACT

Calcium ions mediate a variety of physiological responses of developing neurons including survival. The purpose of this study was to examine the effect of calcium influx through L-type calcium channels (LTCCs) or NMDA receptors on prostaglandin E2 (PGE2)-induced apoptosis in rat cortical cells. Cultures of rat cortical cells were prepared from an embryonic day 18 rat neocortex. After culturing for 2 or 8 days in vitro (DIV), the cells were subjected to PGE2 treatment for 48 h. FPL64176, an LTCC agonist, protected the cells at 2 and 8 DIV from PGE2-induced apoptosis. On the other hand, N-methyl-D-aspartate (NMDA), an agonist of NMDA receptor, protected the cells from PGE2-induced apoptosis only at 8 DIV. FPL64176 increased the calcium levels at 2 and 8 DIV, whereas NMDA increased the calcium levels only at 8 DIV. The protective effects of the LTCC agonist and NMDA on PGE2-induced apoptosis were blocked following treatment of the cells with protein kinase C inhibitors. Our results suggest that LTCCs and NMDA receptors modulate the cell death of developing cortical neurons possibly through a protein kinase C pathway.


Subject(s)
Apoptosis/drug effects , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Calcium/metabolism , Cerebral Cortex/cytology , Dinoprostone/pharmacology , Ions/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Calcium Channel Agonists/pharmacology , Carbazoles/pharmacology , Cells, Cultured , Cerebral Cortex/embryology , Enzyme Inhibitors/pharmacology , N-Methylaspartate/pharmacology , Neurons/drug effects , Protective Agents/pharmacology , Protein Kinase C/antagonists & inhibitors , Pyrroles/pharmacology , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/agonists
2.
Neurosci Res ; 77(1-2): 87-96, 2013.
Article in English | MEDLINE | ID: mdl-23892131

ABSTRACT

Activated microglial cells play an important role in immune and inflammatory responses in CNS and play a role in neurodegenerative diseases. We examined the effects of lipoic acid (LA) on inflammatory responses of BV-2 microglial cells activated by lipopolysaccharide (LPS), and explored the underlying mechanisms of action of LA. BV-2 cells treated with LPS showed an up-regulation of mRNA of the pro-inflammatory molecules, inducible nitric oxide synthase (iNOS). LA suppressed the expression of iNOS and furthermore, LPS-induced production of nitrite. Moreover, LA suppressed the nuclear translocation of RelA, a component of nuclear factor-kappa B (NF-κB) that contains transcriptional activator domain for LPS. The mechanisms of LA-mediated anti-inflammatory effects on microglia remain unknown, and we suggested an involvement of Akt/glycogen synthase kinase-3ß (GSK-3ß) phosphorylation. The results showed that inhibitor of phosphatidylinositol 3-kinase prevented LA-mediated suppression of LPS induction of RelA and expression of iNOS. Furthermore, these inflammatory actions were prevented by GSK-3ß inhibitors. These data demonstrate a role for LA as a chemical modulator of inflammatory responses by microglia, and thus may be a therapeutic strategy for treating neurodegenerative diseases with an inflammatory component.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Glycogen Synthase Kinase 3/metabolism , Microglia/drug effects , Microglia/enzymology , Nitric Oxide Synthase Type II/metabolism , Thioctic Acid/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Lipopolysaccharides/pharmacology , Lithium Chloride/pharmacology , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor RelA/metabolism
3.
Neurochem Int ; 61(7): 1242-53, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22995787

ABSTRACT

Nipradilol (Nip), which has α1- and ß-adrenoceptor antagonist and nitric oxide (NO)-donating properties, has clinically been used as an anti-glaucomatous agent in Japan. NO mediates cellular signaling pathways that regulate physiological functions. The major signaling mechanisms mediated by NO are cGMP-dependent signaling and protein S-nitrosylation-dependent signalings. Nip has been described as having neuroprotective effects through cGMP-dependent pathway in retinal ganglion cells (RGCs). However, the effect seems to be partial. On the other hand, whether Nip can prevent cell death through S-nitrosylation is not yet clarified. In this study, we therefore focused on the neuroprotective mechanism of Nip through S-nitrosylation. Nip showed a dramatic neuroprotective effect against oxidative stress-induced death of RGC-5 cells. However, denitro-nipradilol, which does not have NO-donating properties, was not protective against oxidative stress. Furthermore, an NO scavenger significantly reversed the protective action of Nip against oxidative stress. In addition, we demonstrated that α1- or ß-adrenoceptor antagonists (prazosin or timolol) did not show any neuroprotective effect against oxidative stress in RGC-5 cells. We also demonstrated that Nip induced the expression of the NO-dependent antioxidant enzyme, heme oxygenase-1 (HO-1). S-nitrosylation of Kelch-like ECH-associated protein by Nip was shown to contribute to the translocation of NF-E2-related factor 2 to the nucleus, and triggered transcriptional activation of HO-1. Furthermore, RGC death and levels of 4-hydroxy-2-nonenal (4HNE) were increased after optic nerve injury in vivo. Pretreatment with Nip significantly suppressed RGC death and accumulation of 4HNE after injury through an HO-1 activity-dependent mechanism. These data demonstrate a novel neuroprotective action of Nip against oxidative stress-induced RGC death in vitro and in vivo.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Heme Oxygenase-1/biosynthesis , Nitric Oxide/metabolism , Propanolamines/pharmacology , Retinal Ganglion Cells/drug effects , Vasodilator Agents/pharmacology , Animals , Base Sequence , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , DNA Primers , Enzyme Induction , Immunohistochemistry , Kelch-Like ECH-Associated Protein 1 , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Retinal Ganglion Cells/enzymology , Retinal Ganglion Cells/metabolism
4.
Neurotox Res ; 21(3): 309-16, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22006118

ABSTRACT

In the central nervous system, members of the Src family of tyrosine kinases (SFKs) are widely expressed and are abundant in neurons. The purpose of this study is to examine whether glycogen synthase-3 (GSK-3) is involved in SFK inhibitor-induced apoptosis. PP2 and SU6656, SFK inhibitors, increased apoptotic cell death with morphological changes that were characterized by cell shrinkage, chromatin condensation, or nuclear fragmentation. Moreover, both activation of caspase-9 and caspase-3 were accompanied by the cell death. GSK-3 inhibitors, such as alsterpaullone and SB216763, prevented the PP2-induced apoptosis. In addition, insulin-like growth factor-I prevented the PP2-induced cell death and PP2 inhibited phosphorylation of focal adhesion kinase (FAK). Phosphorylation of FAK on Tyr 576 by Src activates FAK. These results suggest that inhibition of SFK induces apoptosis possibly via blocking of FAK/phosphatidylinositol-3 kinase/Akt signaling pathway and activation of GSK-3 is involved in the cell death in rat cortical neurons.


Subject(s)
Apoptosis/drug effects , Indoles/pharmacology , Neurons/drug effects , Neurons/enzymology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Apoptosis/physiology , Benzazepines/pharmacology , Caspase 3/metabolism , Caspase 9/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Enzyme Inhibitors/pharmacology , Female , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Insulin-Like Growth Factor I/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Maleimides/pharmacology , Neurons/cytology , Neuroprotective Agents/pharmacology , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , src-Family Kinases/metabolism
5.
Neurotox Res ; 20(1): 26-31, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20809179

ABSTRACT

Iron accumulation in brain is associated with a number of common neurodegenerative disorders. N-ß-alanyl-5-S-glutathionyl-3,4-dihydroxyphenylalanine (5-S-GAD), a novel catechol derivative, was isolated from adult flesh fly as a defense substance. We examined the effect of 5-S-GAD on Fe²+-induced lipid peroxidation and cell death in PC12 cells. Treatment of PC12 cells with Fe²+ (1-20 µM) for 24 h induced lipid peroxidation and cell death in a dose-dependent manner. Butylated hydroxyanisole and α-tocopherol inhibited Fe²+-induced lipid peroxidation and cell death. 5-S-GAD inhibited Fe²+-induced lipid peroxidation and cell death. 5-S-GAD protected PC12 cells from Fe²+-induced cell death possibly by blocking lipid peroxidation.


Subject(s)
Cell Death/drug effects , Dihydroxyphenylalanine/analogs & derivatives , Ferrous Compounds/antagonists & inhibitors , Glutathione/analogs & derivatives , Lipid Peroxidation/drug effects , Neurons/physiology , Animals , Butylated Hydroxyanisole/pharmacology , Cell Death/physiology , Dihydroxyphenylalanine/pharmacology , Dose-Response Relationship, Drug , Ferrous Compounds/pharmacology , Glutathione/pharmacology , Neurons/drug effects , PC12 Cells , Rats , Thiobarbituric Acid Reactive Substances/metabolism , Tocopherols/pharmacology
6.
Cell Mol Neurobiol ; 30(6): 863-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20300959

ABSTRACT

The purpose of this study was to examine whether glycogen synthase kinase-3 (GSK-3) is involved in colchicine-induced cell death in PC12 cells by using GSK inhibitors. Colchicine increased apoptotic cell death with morphological changes characterized by cell shrinkage and nuclear condensation or fragmentation. GSK-3 inhibitors such as alsterpaullone, SB216763, and AR-A014418 prevented colchicine-induced cell death and caspase-3 activation. These results suggest that colchicine induces caspase-dependent apoptotic cell death and that GSK-3 activation is involved in cell death in PC12 cells.


Subject(s)
Apoptosis/drug effects , Colchicine/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Caspase 3/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3/metabolism , PC12 Cells , Phosphorylation/drug effects , Protective Agents/pharmacology , Rats
7.
Cell Mol Neurobiol ; 30(2): 193-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19688259

ABSTRACT

Calcium ion is a secondary messenger that mediates a variety of physiological responses of neurons, including cell survival responses. To determine the role of calcium in regulating neuronal survival and death, we examined whether chelation of extracellular calcium with EGTA induces caspase-dependent apoptotic cell death and whether glycogen synthase kinase-3 is involved in EGTA-induced cell death in PC12 cells. EGTA increased apoptotic cell death with morphological changes characterized by cell shrinkage and nuclear condensation and fragmentation accompanied by caspase activation. EGTA increased GRP78 protein expression, suggesting that EGTA induces ER stress. Glycogen synthase kinase-3 inhibitors prevented EGTA-induced apoptosis. In addition, nerve growth factor and insulin growth factor-I completely blocked EGTA-induced cell death. Moreover, caspase-3 activation was inhibited by glycogen synthase kinase-3 inhibitors. These results suggest that chelation of extracellular calcium with EGTA induces caspase-dependent apoptosis, and the activation of glycogen synthase kinase-3 is involved in the death of PC12 cells.


Subject(s)
Calcium , Cell Death/drug effects , Chelating Agents/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Animals , Apoptosis/physiology , Benzazepines/metabolism , Calcium/metabolism , Calcium/pharmacology , Caspases/metabolism , Egtazic Acid/metabolism , Enzyme Activation , Heat-Shock Proteins/metabolism , Indoles/metabolism , Insulin-Like Growth Factor I/metabolism , Maleimides/metabolism , Nerve Growth Factor/metabolism , PC12 Cells , Rats
8.
J Biomed Sci ; 15(6): 767-70, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18712491

ABSTRACT

The purpose of this study is to examine whether benzyl alcohol affects N-methyl-D-aspartate (NMDA) receptor in cortical cells. Benzyl alcohol (0.5-2 mM) inhibited NMDA-induced cytotoxicity. The protective effect of benzyl alcohol on NMDA-induced toxicity disappeared by washing cells with buffer to remove benzyl alcohol. Benzyl alcohol reduced NMDA receptor-mediated calcium accumulation, indicating that benzyl alcohol inhibits NMDA receptor activity.


Subject(s)
Anesthetics, Local/pharmacology , Benzyl Alcohol/pharmacology , Calcium/metabolism , Cerebral Cortex/drug effects , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cells, Cultured , Excitatory Amino Acid Agonists/toxicity , N-Methylaspartate/toxicity , Rats , Time Factors
9.
Cell Mol Neurobiol ; 27(6): 783-90, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17647100

ABSTRACT

Calmodulin is known to transduce Ca(2+) signals by interacting with specific target proteins. In order to determine the role of calmodulin in regulating neuronal survival and death, we examined, whether calmodulin inhibitors induce caspase-dependent apoptotic cell death, and whether glycogen synthase kinase-3 is involved in calmodulin inhibitor-induced cell death in PC12 cells. W13, a calmodulin specific inhibitor increased apoptotic cell death with morphological changes characterized by cell shrinkage and nuclear condensation of fragmentation. Glycogen synthase kinase-3 inhibitors prevented calmodulin inhibitor-induced apoptosis. In addition, nerve growth factor and cycloheximide, a protein synthesis inhibitor, completely blocked cell death. Moreover, caspase-3 activation was accompanied by calmodulin inhibitor-induced cell death and inhibited by nerve growth factor. These results suggest that calmodulin inhibitors induce caspase-dependent apoptosis, and the activation of glycogen synthase kinase-3 is involved in the death of PC12 cells.


Subject(s)
Apoptosis/drug effects , Benzazepines/pharmacology , Calmodulin/antagonists & inhibitors , Glycogen Synthase Kinase 3/antagonists & inhibitors , Indoles/pharmacology , Maleimides/pharmacology , Sulfonamides/pharmacology , Animals , Caspase 3/metabolism , Cell Death/drug effects , L-Lactate Dehydrogenase/metabolism , PC12 Cells , Protein Kinase Inhibitors/pharmacology , Rats
10.
Neurosci Lett ; 423(1): 24-8, 2007 Aug 09.
Article in English | MEDLINE | ID: mdl-17640805

ABSTRACT

Hypothermia protects against hypoxic or ischemic damage. However, the mechanisms by which brain cooling prevents hypoxic or ischemic damage are not clear. We examined whether hypothermia protects against excitotoxicity in cultured cortical cells. Exposure of cortical cell culture to 500 microM N-methyl-D-aspartate (NMDA) for 15 min at 32 degrees C or 37 degrees C did not induce neurotoxicity. On the other hand, reduction of temperature to 20 degrees C resulted in widespread neuronal disintegration by the following day. Moreover, intracellular calcium concentration increased markedly by adding NMDA to cells at 20 degrees C. These results suggest that profound hypothermia does not protect neurons from excitotoxicity by inhibiting NMDA receptor activity.


Subject(s)
Cell Survival/physiology , Cerebral Cortex/physiology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Calcium/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Fluorescent Dyes , Hypothermia, Induced , L-Lactate Dehydrogenase/metabolism , Neuroglia/drug effects , Neurons/drug effects , Pregnancy , Rats , Temperature
11.
Neurochem Res ; 32(8): 1336-42, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17401651

ABSTRACT

Calcium ion is essential for cellular functions including signal transduction. Uncontrolled calcium stress has been linked causally to a variety of neurodegenerative diseases. Thapsigargin, which inhibits Ca(2+)-ATPase in the endoplasmic reticulum (ER) and blocks the sequestration of calcium by the ER, induced apoptotic cell death (chromatin condensation and nuclear fragmentation) accompanied by GRP78 protein expression and caspase-3 activation in rat fetal cortical neurons (days in vitro 9-10). Blockade of N-methyl-D-aspartate (NMDA) receptors with NMDA antagonists induced apoptosis without GRP78 protein expression. Apoptosis accompanied both caspase-9 and caspase-3 activation. We then examined whether GSK-3 is involved in thapsigargin-induced cell death by using GSK-3 inhibitors. We assayed the effects of selective GSK-3 inhibitors, SB216763, alsterpaullone and 1-azakenpaullone, on thapsigargin-induced apoptosis. These inhibitors completely protected cells from thapsigargin-induced apoptosis. In addition, GSK-3 inhibitors inhibited caspase-9 and caspase-3 activation accompanied by thapsigargin-induced apoptosis. These results suggest that thapsigargin induces caspase-dependent apoptosis mediated through GSK-3beta activation in rat cortical neurons.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Cerebral Cortex/cytology , Enzyme Inhibitors/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Neurons/metabolism , Thapsigargin/metabolism , Animals , Cells, Cultured , Dizocilpine Maleate , Enzyme Activation , Excitatory Amino Acid Antagonists/metabolism , Glycogen Synthase Kinase 3/metabolism , Neurons/cytology , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
12.
Brain Res ; 1133(1): 20-6, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17166486

ABSTRACT

Calcineurin is selectively enriched within neurons of the central nervous system. The mechanism of calcineurin inhibitor-induced neurotoxicity remains poorly understood. The purpose of this study is to examine whether glycogen synthase-3 (GSK-3) is involved in calcineurin inhibitor-induced apoptosis. Calcineurin inhibitors such as cyclosporine A (CsA) and FK506 increased apoptotic cell death with morphological changes characterized by cell shrinkage, nuclear condensation of fragmentation, and internucleosomal DNA fragmentation. Alsteropaullone and 1-azakenpaullone, GSK-3 inhibitors, prevented calcineurin inhibitor-induced apoptosis. In addition, insulin growth factor-I (IGF-I) and cycloheximide completely blocked cell death. Moreover, caspase-3 activation was accompanied by calcineurin inhibitor-induced cell death. These results suggest that calcineurin inhibitors induce caspase-dependent apoptosis and activation of GSK-3 is involved in cell death in rat cortical neurons.


Subject(s)
Apoptosis/drug effects , Calcineurin Inhibitors , Caspases/drug effects , Cyclosporine/toxicity , Enzyme Inhibitors/toxicity , Glycogen Synthase Kinase 3/antagonists & inhibitors , Animals , Apoptosis/physiology , Calcineurin/metabolism , Caspases/metabolism , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , DNA Fragmentation/drug effects , Glycogen Synthase Kinase 3/metabolism , Immunosuppressive Agents/toxicity , Insulin-Like Growth Factor I/metabolism , Neurons/drug effects , Neurons/metabolism , Rats , Tacrolimus/toxicity
13.
Neurosci Lett ; 408(2): 124-8, 2006 Nov 13.
Article in English | MEDLINE | ID: mdl-16982147

ABSTRACT

Uncontrolled calcium stress has been linked causally to a variety of neurodegenerative diseases, including ischemia, excitotoxicity and Alzheimer's disease. Thapsigargin, which increases [Ca2+]i, induces apoptotic cell death (chromatin condensation and DNA fragmentation) accompanied by caspase-3 activation in PC12 cells. We examined whether GSK-3 is involved in thapsigargin-induced cell death by using GSK-3 inhibitors in PC12 cells. Cells treated with 0.1 microM thapsigargin for 24h shrank. The injured cells underwent chromatin condensation and nuclear fragmentation, indicating apoptotic cell death. We assayed the effects of selective GSK-3 inhibitors, SB216763, azakenpaullone and alsteropaullone on thapsigargin-induced apoptosis. These inhibitors completely protected cells from thapsigargin-induced apoptosis. Alsterpaullone did not reduce the GRP78 protein expression induced by thapsigargin, suggesting that GSK-3 activation is not involved in induction of GRP78. In addition, GSK-3 inhibitors inhibited caspase-3 activation accompanied by thapsigargin-induced apoptosis. We showed in this report that thapsigargin-induced apoptosis is prevented by GSK-3 inhibitors, suggesting that thapsigargin induces caspase-dependent apoptosis mediated through GSK-3 activation in PC12 cells.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , PC12 Cells/drug effects , Thapsigargin/pharmacology , Animals , Apoptosis/physiology , Benzazepines/pharmacology , Heat-Shock Proteins/metabolism , Indoles/pharmacology , Maleimides/pharmacology , Molecular Chaperones/metabolism , Neuroprotective Agents/pharmacology , Rats
14.
Neurosci Lett ; 400(1-2): 105-9, 2006 May 29.
Article in English | MEDLINE | ID: mdl-16504398

ABSTRACT

Cyclooxygenase-2 (COX-2) induction and prostaglandin E(2) (PGE(2)) elevation have been reported to occur after cerebral ischemic insult. PGE(2) induces apoptosis through the PGE(2) EP2 receptor by a cAMP-dependent pathway. Glycogen synthase kinase-3 (GSK-3) affects many fundamental cellular functions. We examined whether GSK-3 is involved in PGE(2)-induced cell death by using GSK-3 inhibitors in rat cultured cortical neurons. Cells treated with 12.5 microM PGE(2) for 2 days shrank. The injured cells underwent chromatin condensation and nuclear fragmentation detected by staining with Hoechst33258, indicating apoptotic cell death. We assayed the effects of selective GSK-3 inhibitors SB216763 and alsteropaullone on PGE(2)-induced apoptosis. These inhibitors completely protected the cells from apoptosis induced by PGE(2). Moreover, dibutyryl cAMP (a cell permeable cAMP)-induced apoptosis was also prevented by alsteropaullone. In addition, GSK-3 inhibitors inhibited caspase-3 activation accompanied by PGE(2)-induced apoptosis. We showed in this report that PGE(2)-induced apoptosis is prevented by GSK-3 inhibitors, suggesting that PGE(2) induces caspase-dependent apoptosis mediated through GSK-3 activation in rat cultured cortical neurons.


Subject(s)
Apoptosis/drug effects , Cerebral Cortex/cytology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Indoles/pharmacology , Maleimides/pharmacology , Neurons/drug effects , Animals , Benzazepines/pharmacology , Bucladesine/pharmacology , Caspase 3 , Caspases/metabolism , Cell Count/methods , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , In Vitro Techniques , Phosphopyruvate Hydratase/metabolism , Rats , Time Factors
15.
Toxicol Appl Pharmacol ; 210(1-2): 100-7, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16307766

ABSTRACT

Recent data suggest that anesthetic drugs cause neurodegeneration during development. Ketamine is frequently used in infants and toddlers for elective surgeries. The purpose of this study is to determine whether glycogen synthase kinase-3 (GSK-3) is involved in ketamine-induced apoptosis. Ketamine increased apoptotic cell death with morphological changes which were characterized by cell shrinkage, nuclear condensation or fragmentation. In addition, insulin growth factor-1 completely blocked the ketamine-induced apoptotic cell death. Ketamine decreased Akt phosphorylation. GSK-3 is known as a downstream target of Akt. The selective inhibitors of GSK-3 prevented the ketamine-induced apoptosis. Moreover, caspase-3 activation was accompanied by the ketamine-induced cell death and inhibited by the GSK-3 inhibitors. These results suggest that activation of GSK-3 is involved in ketamine-induced apoptosis in rat cortical neurons.


Subject(s)
Anesthetics, Dissociative/adverse effects , Apoptosis/drug effects , Cerebral Cortex/drug effects , Ketamine/adverse effects , Neurons/drug effects , Animals , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/enzymology , Dizocilpine Maleate/pharmacology , Glycogen Synthase Kinase 3/metabolism , N-Methylaspartate/pharmacology , Neurons/cytology , Neurons/enzymology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar
16.
Life Sci ; 78(16): 1878-83, 2006 Mar 13.
Article in English | MEDLINE | ID: mdl-16309709

ABSTRACT

The activation of glutamate receptors, particularly N-methyl-D-aspartate (NMDA) receptors, initiates ischemic cascade in the early stages of cerebral ischemia. Postischemia, cerebral ischemia is also associated with an inflammatory reaction that contributes to tissue damage. The up-regulation of neuronal cyclooxygenase-2 (COX-2) and elevation of prostaglandin E2 (PGE2) have been reported to occur after cerebral ischemic insult. We therefore studied whether the COX-2 reaction product PGE2 affects glutamate receptor-mediated cell death in cultured rat cortical cells. PGE2 was found to augment NMDA-mediated cell death. The transcription of EP1, EP2, EP3 and EP4 PGE2 receptor genes was investigated using reverse transcriptase-polymerase chain reaction (RT-PCR). EP1, EP2 and EP3 receptor genes were found in cortical cells. Butaprost (an EP2 agonist) markedly enhanced NMDA-mediated cell death, whereas 17-phenyl trinor-PGE2 (an EP1 agonist) and sulprostone (an EP3 agonist) had little effect. Both PGE2 and butaprost elevated cAMP intracellular levels in the cortical cells; moreover, forskolin, an activator of adenylate cyclase, enhanced NMDA-mediated cell death. These results suggest that PGE2, acting via EP2 receptors, aggravates excitotoxic neurodegeneration by a cAMP-dependent mechanism.


Subject(s)
Apoptosis/drug effects , Cerebral Cortex/drug effects , Dinoprostone/pharmacology , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Prostaglandin E/metabolism , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cyclic AMP/metabolism , Female , Fetus/drug effects , Neurons/metabolism , Neurons/pathology , Pregnancy , Prostaglandins E, Synthetic/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP2 Subtype , Reverse Transcriptase Polymerase Chain Reaction
17.
Biol Pharm Bull ; 28(6): 941-6, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15930723

ABSTRACT

Exposure of rat phenochromocytoma cells (PC12 cells) to aluminum maltolate complex, Al(maltol)3, induced a decrease in intracellular glutathione (GSH) concentration, resulting in a facilitated release of lactate dehydrogenase (LDH) from the cell and an increase in trypan blue-stained cells. Similar phenomena were observed as the cells were treated with L-buthione-[S,R]-sulfoximine (BSO) in the presence of Al(maltol)3. On the other hand, treatment of PC 12 cells with BSO alone in the absence of Al(maltol)3 did not affect the cell viability. Pre-treatment of PC12 cells with N-acetylcysteine (NAC) for 30 min before a 48 h-exposure to Al(maltol)3 effectively protected the cells from Al(maltol)3 toxicity by increasing intracellular GSH concentration. NAC also effectively inhibited reactive oxygen species (ROS) generation induced by treatment of the cells with Al(maltol)3. However, several lipophilic radical scavengers such as alpha-tocopherol and 3(2)-tert-butyl-4-hydroxyanisole, and an iron chelator, desferrioxamine, did not prevent Al(maltol)3-mediated ROS production or the decrease of cell viability. Based on these results, we discussed the role of intracellular GSH against the onset of aluminum toxicity in the context of ROS production.


Subject(s)
Glutathione/deficiency , Organometallic Compounds/toxicity , Pyrones/toxicity , Animals , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , PC12 Cells , Rats , Reactive Oxygen Species/metabolism
18.
Eur J Pharmacol ; 499(3): 239-45, 2004 Sep 24.
Article in English | MEDLINE | ID: mdl-15381045

ABSTRACT

The effect of ethanol on cell viability was examined in rat cultured cortical neurons. Ethanol induced apoptosis, which was characterized by cell shrinkage, nuclear condensation or fragmentation and internucleosomal DNA fragmentation. Ethanol-induced apoptosis was prevented by N-methyl-d-aspartate (NMDA), an agonist of the NMDA receptor, which is a subtype of ionotropic glutamate receptors. Incubation with glycogen synthase kinase-3 (GSK-3) inhibitors 3-(2,4-dichlorophenyl)-4-(1-methyl-1H-indol-3-yl)-1H-pyrrole-2,5-dione (SB216763) and alsteropaullone, but not a cyclin-dependent protein kinase 5 inhibitor roscovitine, completely protected the neurons from ethanol-induced apoptosis. Apoptosis was accompanied by the activation of caspase-3 and prevented by a caspase-3 inhibitor. These results suggest that ethanol induces caspase-dependent apoptosis mediated by glycogen synthase kinase-3 activation in cultured rat cortical neurons.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Cerebral Cortex/drug effects , Ethanol/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Calcium/metabolism , Caspase 3 , Caspase Inhibitors , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Indoles/pharmacology , Intracellular Space/drug effects , Intracellular Space/metabolism , Maleimides/pharmacology , N-Methylaspartate/pharmacology , Peptides , Proteins/pharmacology , Purines/pharmacology , Rats , Roscovitine
19.
Brain Res ; 1020(1-2): 196-203, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15312803

ABSTRACT

The N-methyl-d-aspartate (NMDA) receptor 2B-selective antagonist ifenprodil induced morphological changes which were characterized by cell shrinkage, nuclear condensation or fragmentation, and internucleosomal DNA fragmentation in rat cultured cortical cells. Ifenprodil increased the apoptotic cell death in a dose-dependent manner (0.5-10 microM). In addition, the protein synthesis inhibitor cycloheximide completely blocked ifenprodil-induced apoptotic cell death. The selective inhibitors of glycogen synthase kinase-3 (GSK-3) prevented the ifenprodil-induced apoptosis. Moreover, activation of caspase-3 was accompanied by cell death induced by ifenprodil in a dose-dependent manner. The ifenprodil-induced apoptosis was prevented by a caspase-3 inhibitor. These results suggested that activation of GSK-3 involves in the apoptosis induced by blocking of trophic effect of NMDA receptor consisting of NR2B subunit in rat cortical neurons.


Subject(s)
Apoptosis/drug effects , Cerebral Cortex/metabolism , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/drug effects , Neurons/drug effects , Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Caspases/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3/metabolism , Neurons/enzymology , Rats
20.
Neurochem Int ; 45(5): 713-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15234114

ABSTRACT

Cyclooxygenase-2 (COX-2) induction and prostaglandin E2 elevation have been reported to occur after cerebral ischemic insult. To evaluate whether the cyclooxygenase-2 reaction product prostaglandin E2 is directly related to induction of apoptosis in neuronal cells, the effect of prostaglandin E2 on cell viability was examined in hippocampal cells. Prostaglandin E2 (5-25 microM) induced apoptosis in a dose-dependent manner 48 h after addition to the cells, which was characterized by cell shrinkage, nuclear condensation or fragmentation and attenuated by a protein synthesis inhibitor, cycloheximide. Neither 17-phenyl trinor-prostaglandin E2 (an EP1 agonist) nor sulprostone (an EP3 agonist) induced cell death, whereas butaprost (an EP2 agonist) induced apoptosis. Prostaglandin E2 increased the intracellular concentration of cAMP, and the selective EP2 agonist butaprost also induced apoptosis accompanied by increasing cAMP levels in hippocampal cells. Moreover, a cell permeable cAMP analog, dibutyryl cAMP also induced apoptosis in hippocampal cells. These findings suggest that prostaglandin E2-induced apoptosis was mediated through a mechanism involving the cAMP-dependent pathway. In addition, prostaglandin E2 activated caspase-3 activity in a dose-dependent manner and a caspase-3 inhibitor prevented the prostaglandin E2-induced apoptosis. We showed in this report that prostaglandin E2 directly induced apoptosis in hippocampal neurons. Moreover, it is likely that the direct effects of prostaglandin E2 on hippocampal neurons were mediated by activation of EP2 receptors followed by elevation of the intracellular cAMP levels.


Subject(s)
Alprostadil/analogs & derivatives , Apoptosis/drug effects , Caspases/physiology , Dinoprostone/toxicity , Hippocampus/cytology , Hippocampus/drug effects , Neurons/drug effects , Receptors, Prostaglandin E/agonists , Alprostadil/pharmacology , Animals , Bucladesine/pharmacology , Caspase 3 , Caspases/metabolism , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cells, Cultured , Cyclic AMP/metabolism , Cycloheximide/pharmacology , DNA Primers , Dose-Response Relationship, Drug , Female , Pregnancy , Protein Synthesis Inhibitors/pharmacology , Rats , Receptors, Prostaglandin E/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...