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










Publication year range
1.
Eur J Neurol ; 18(9): 1151-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21299736

ABSTRACT

BACKGROUND AND PURPOSE: Extensive evidence has shown that oxidative stress mediates neuronal death in animal models of hypoxic-ischaemia. Brain biomarkers of oxidative stress need to be identified in order to better understand and treat brain damage in human stroke patients. The present study was conducted to identify potential target proteins of oxidative stress in the cerebrospinal fluid (CSF) of stroke patients with acute ischaemic brain injury. METHODS: We performed two-dimensional polyacrylamide gel electrophoresis to separate protein samples obtained from the CSF of control and stroke patients. To determine protein oxidation levels, oxyblot was then used to detect protein carbonyls that were determined by formation of a stable 2,4-dinitrophenylhydrazine (DNP) product using an anti-DNP antibody. RESULTS: We found that oxidation of serum albumin was increased in the CSF from stroke patients as well as rats who underwent permanent middle cerebral artery occlusion (6.5%, 23%, respectively). In stroke patients, oxidized albumin levels correlated to neurologic indications. CONCLUSIONS: The present study suggests that oxidized albumin in CSF can be utilized as an oxidative stress marker in human stroke patients.


Subject(s)
Biomarkers/cerebrospinal fluid , Oxidative Stress/physiology , Serum Albumin/cerebrospinal fluid , Stroke/cerebrospinal fluid , Animals , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Male , Middle Aged , Rats , Rats, Sprague-Dawley
2.
J Neurochem ; 85(2): 525-33, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12675929

ABSTRACT

Blockade of ionotropic glutamate receptors induces neuronal cell apoptosis. We investigated if mitochondria-mediated death signals would contribute to neuronal apoptosis following administration of glutamate antagonists. The administration of MK-801 and CNQX (MK-801/CNQX), the selective antagonists of N-methyl-d-aspartate (NMDA) and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/kainate receptors, produced widespread neuronal death in neonatal rat brain and cortical cell cultures. MK-801/CNQX-induced neuronal apoptosis was prevented by zVAD-fmk, a broad inhibitor of caspases, but insensitive to inhibitors of calpain or cathepsin D. Activation of caspase-3 was observed within 6-12 h and sustained over 36 h after exposure to MK-801/CNQX, which cleaved PHF-1 tau, the substrate for caspase-3. Activation of caspase-3 was blocked by high K+ and mimicked by BAPTA-AM, a selective Ca2+ chelator. Reducing extracellular Ca2+, but not Na+, activated caspase-3, suggesting an essential role of Ca2+ deficiency in MK-801/CNQX-induced activation of caspases. Cortical neurons treated with MK-801/CNQX triggered activation of caspase-9, release of cytochrome c from mitochondria, and translocation of Bax into mitochondria. The present study suggests that blockade of ionotropic glutamate receptors causes caspase-3-mediated neuronal apoptosis due to Ca2+ deficiency that is coupled to the sequential mitochondrial death pathway.


Subject(s)
Apoptosis/physiology , Calcium/deficiency , Neurons/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2 , Receptors, Glutamate/drug effects , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Apoptosis/drug effects , Calcium/metabolism , Caspase 3 , Caspase 9 , Caspase Inhibitors , Caspases/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Chelating Agents/pharmacology , Cytochrome c Group/metabolism , Dizocilpine Maleate/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Mice , Neurons/cytology , Potassium/pharmacology , Protein Transport/drug effects , Proto-Oncogene Proteins/metabolism , Rats , Rats, Sprague-Dawley , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/metabolism , Receptors, Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , bcl-2-Associated X Protein
3.
Neurobiol Dis ; 8(5): 774-83, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11592847

ABSTRACT

Synaptically released Zn2+ ions enter into neurons primarily through voltage-gated Ca2+ channels (VGCC) or N-methyl-d-aspartate (NMDA) receptors, which can mediate pathological neuronal death. We studied the possibility (and underlying mechanisms) that aspirin, known to prevent NMDA neurotoxicity, would also attenuate Zn2+ neurotoxicity. Administration of 3 to 10 mM aspirin, in cortical cell cultures, attenuated the evolution of neuronal death following exposure to 300 microM Zn2+ for 30 min. This neuroprotective effect of aspirin was attributable to the prevention of Zn2+ ion entry. Aspirin interfered with inward currents and an increase in [Ca2+]i through VGCC and selective binding of omega-conotoxin, sensitive to N-type Ca2+ channel. The omega-conotoxins GVIA or MVIIC, the selective inhibitors of N-type Ca2+ channels, attenuated Zn2+ neurotoxicity. Aspirin derivatives lacking the carboxyl acid group did not reduce Zn2+ neurotoxicity. The present findings suggest that aspirin prevents Zn2+-mediated neuronal death by interfering with VGCC, and its action specifically requires the carboxyl acid group.


Subject(s)
Apoptosis/drug effects , Aspirin/pharmacology , Calcium Channels, N-Type/physiology , Ion Transport/drug effects , Nerve Tissue Proteins/physiology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Zinc/toxicity , Acetylcysteine/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Aspirin/analogs & derivatives , Aspirin/chemistry , Benzoates/pharmacology , Calcium/metabolism , Calcium Channels, N-Type/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Chromans/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , Mice , N-Methylaspartate/pharmacology , Nerve Tissue Proteins/drug effects , Neurons/metabolism , Staurosporine/pharmacology , Structure-Activity Relationship , Zinc/antagonists & inhibitors , Zinc/pharmacology , omega-Conotoxin GVIA/pharmacology , omega-Conotoxins/pharmacology
4.
J Pharmacol Exp Ther ; 299(1): 377-84, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11561102

ABSTRACT

Complestatin, a peptide derived from Streptomyces, was found to protect cultured cortical neurons from excitotoxicity induced by N-methyl-D-aspartate (NMDA), alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), or kainate. This neuroprotective behavior of complestatin was attributed to a blockade of Ca2+ ion entry and accumulation, after the activation of NMDA and AMPA/kainate receptors. Complestatin reversibly interfered with NMDA- and AMPA-mediated excitatory synaptic transmission. Complestatin also protected cortical neurons from prolonged deprivation of oxygen and glucose, more effectively than combined antagonists of NMDA and AMPA/kainate receptors. Neurotoxicity, evolving within 1 to 2 days after continuous exposure to combined NMDA and AMPA/kainate antagonists, was not observed in cortical cell cultures that were exposed to complestatin. Finally, complestatin dose dependently prevented neuronal death evolving within the inner nuclear and ganglion cell layers, after transient retinal ischemia. We conclude that complestatin possesses novel pharmacological properties that effectively prevent excitotoxicity under certain pathological conditions.


Subject(s)
Brain Ischemia/pathology , Chlorophenols/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Peptides, Cyclic , Receptors, AMPA/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Apoptosis/drug effects , Calcium/metabolism , Cell Death/drug effects , Excitatory Amino Acid Agonists/toxicity , Glucose/deficiency , Kainic Acid/antagonists & inhibitors , Kainic Acid/toxicity , Mice , Oxidative Stress/drug effects , Oxygen/physiology , Patch-Clamp Techniques , Retinal Vessels/drug effects , Retinal Vessels/physiology
5.
J Neurochem ; 78(2): 230-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11461958

ABSTRACT

Sustained alteration in [Ca(2+)]i triggers neuronal death. We examined morphological and signaling events of Ca(2+)-deficiency-induced neuronal death. Cortical cell cultures exposed to 20 microM 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA-AM), an intracellular calcium chelator, underwent neuronal apoptosis within 12 h that was evident by shriveled cell bodies, aggregated and condensed nuclear chromatin, and disrupted nuclear membrane. Thereafter, surviving neurons revealed typical necrosis, accompanied by swelling of cell body and mitochondria, over 24 h. Both apoptosis and necrosis were prevented by inclusion of 1 microg/mL cycloheximide, a protein synthesis inhibitor. Treatment with BAPTA-AM induced translocation of Bax into mitochondria within 4 h and release of cytochrome c from mitochondria over 4-12 h. An active fragment of caspase-3, a downstream mediator of cytochrome c, was observed within 8 h and cleaved PHF-1-positive tau. Administration of zVAD-fmk, a broad inhibitor of caspases, or DEVD-amc, a selective inhibitor of caspase-3, selectively prevented the apoptosis component of BAPTA-AM neurotoxicity. In contrast, BAPTA-AM-induced necrosis was propagated through sequential production of superoxide, mitochondrial and cytoplasmic reactive oxygen species. Combined treatment with caspase inhibitors and antioxidants blocked BAPTA-AM neurotoxicity. The present study suggests that neurons deficient in [Ca(2+)]i undergo caspase-3-mediated apoptosis and reactive oxygen species (ROS)-mediated necrosis.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Cerebral Cortex/cytology , Egtazic Acid/pharmacology , Neuroglia/cytology , Neurons/cytology , Neurons/physiology , Reactive Oxygen Species/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/physiology , Chelating Agents/pharmacology , Chromans/pharmacology , Cycloheximide/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Dizocilpine Maleate/pharmacology , Egtazic Acid/analogs & derivatives , Fetus , Kinetics , Mice , Mice, Inbred ICR , Necrosis , Neocortex/cytology , Neocortex/physiology , Neuroglia/drug effects , Neuroglia/physiology , Neurons/ultrastructure , Neuroprotective Agents/pharmacology , Time Factors
6.
Mol Cell Neurosci ; 17(6): 1025-33, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11414791

ABSTRACT

Striatal and cortical neurons containing NADPH-diaphorase [NADPH-d(+)] are highly vulnerable to excitotoxicity that is induced by activation of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)- or kainate-sensitive glutamate receptors. This has been attributed to Ca2+ entry through AMPA/kainate receptors in NADPH-d(+) neurons. In this study, we applied single cell RT-PCR technique to test the hypothesis that differences in levels and processing of the GluR2 subunit would contribute to the selective vulnerability of NADPH-d(+) neurons to AMPA. The nested PCR specific for GluR1-GluR4 showed that rat striatal NADPH-d(+) neurons expressed twice as much GluR1 mRNA as NADPH-d(-) neurons did. The percentage of RNA editing at the Q/R site of GluR2 was 46% in NADPH-d(+) neurons and 92% in NADPH-d(-) neurons. These results suggest that the unedited expression of GluR2 and the reduced ratio of GluR2/GluR1 render NADPH-d(+) neurons highly sensitive to Ca2+-mediated AMPA neurotoxicity. In support of this, most NADPH-d(+) neurons exposed to 100 microM AMPA showed Co2+ uptake and survived AMPA challenge only in the absence of extracellular Ca2+.


Subject(s)
NADPH Dehydrogenase/metabolism , Neurons/metabolism , Neurotoxins/pharmacology , RNA Editing/physiology , RNA, Messenger/metabolism , Receptors, AMPA/genetics , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cobalt/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Fetus , Gene Expression/drug effects , Gene Expression/physiology , Neostriatum/cytology , Neostriatum/drug effects , Neostriatum/metabolism , Neurons/cytology , Neurons/drug effects , RNA Editing/drug effects , RNA, Messenger/drug effects , Rats , Receptors, AMPA/agonists , Receptors, AMPA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
7.
Mol Pharmacol ; 59(5): 981-6, 2001 May.
Article in English | MEDLINE | ID: mdl-11306679

ABSTRACT

Zn(2+), one of the most abundant trace metal ions in mammalian cells, modulates the functions of many regulatory proteins associated with a variety of cellular activities. In the central nervous system, Zn(2+) is highly localized in the cerebral cortex and hippocampus. It has been proposed to play a role in normal brain function as well as in the pathophysiology of certain neurodegenerative disorders. We here report that Zn(2+) induced stimulation of the c-Jun N-terminal kinase (JNK) pathway in mouse primary cortical cells and in various cell lines. Exposure of cells to Zn(2+) resulted in the stimulation of JNK and its upstream kinases including stress-activated protein kinase kinase and mitogen-activated protein kinase kinase kinase. Zn(2+) also induced stimulation of phosphoinositide 3-kinase (PI3K) The Zn(2+)-induced JNK stimulation was blocked by LY294002, a PI3K inhibitor, or by a dominant-negative mutant of PI3Kgamma. Furthermore, overexpression of Rac1N17, a dominant negative mutant of Rac1, suppressed the Zn(2+)- and PI3Kgamma-induced JNK stimulation. The stimulatory effect of Zn(2+) on both PI3K and JNK was repressed by the free-radical scavenging agent N-acetylcysteine. Taken together, our data suggest that Zn(2+) induces stimulation of the JNK signaling pathway through PI3K-Rac1 signals and that the free-radical generation may be an important step in the Zn(2+) induction of the JNK stimulation.


Subject(s)
MAP Kinase Kinase 4 , MAP Kinase Kinase Kinase 1 , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Zinc/pharmacology , Acetylcysteine/pharmacology , Animals , Drug Interactions , Enzyme Activation , Free Radical Scavengers/pharmacology , Genes, Reporter , JNK Mitogen-Activated Protein Kinases , Luciferases , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Cells, Cultured
8.
Neuroreport ; 12(5): 963-6, 2001 Apr 17.
Article in English | MEDLINE | ID: mdl-11303769

ABSTRACT

The effects of 5-hydroxytryptamine (5-HT) on several types of neuronal injury in mouse cortical cell cultures were tested. Co-treatment with 5-HT prevented free radical-mediated neuronal necrosis induced by FeCl2 or buthionine sulfoximine (BSO) in a dose-dependent manner. Subtype antagonists did not reverse the protective effect and 5-HT showed direct free radical scavenging activity evidenced by its ability to reduce the stable free radical 1,1-diphenyl-2-picrylhydrazyl (DPPH) in a cell-free system. Excitotoxic necrosis induced by NMDA or apoptosis induced by staurosporine was not sensitive to 5-HT treatment. These features raise the possibility that the endogenous neurotransmitter 5-HT may work as an innate antioxidant defense mechanism in the CNS.


Subject(s)
Antioxidants/pharmacology , Cerebral Cortex/pathology , Free Radicals/toxicity , Neurons/drug effects , Serotonin/pharmacology , Animals , Apoptosis/drug effects , Cell Death/drug effects , Cells, Cultured , Free Radical Scavengers/pharmacology , Mice , Neuroprotective Agents/pharmacology , Serotonin Antagonists/pharmacology , Staurosporine/toxicity
9.
Neurobiol Dis ; 8(1): 162-72, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11162250

ABSTRACT

Using an in vitro translation assay to screen a human brain cDNA library, we isolated the microtubule-associated protein Tau and determined it to be a caspase-3 substrate whose C-terminal cleavage occurred during neuronal apoptosis. DeltaTau, the 50-kDa cleavage product, was detected by Western blot in apoptotic cortical cells probed with anti-PHF-1 and anti-Tau-5 antibodies, but not anti-T-46 antibody which recognizes the C-terminus. Overexpression of DeltaTau in SK-N-BE2(C) cells significantly increased the incidence of cell death. Staurosporine-induced Tau cleavage was blocked by 20 microM z-Asp-Glu-Val-Asp-chloromethylketone, a caspase-3 inhibitor, and in vitro, Tau was selectively cleaved by caspase-3 or calpain, a calcium-activated protease, but not by caspases-1, -8, or -9. (D421E)-Tau, a mutant in which Asp421 was replaced with a Glu, was resistant to cleavage by caspase-3 and tended to suppress staurosporine-induced cell death more efficiently than did wild-type Tau in both transient and stable expression systems. Finally, the incidence of DeltaTau-induced cell death was augmented by expression of Abeta precursor protein (APP) or Swedish APP mutant. Taken together, these results suggest that the caspase-3 cleavage product of Tau may contribute to the progression of neuronal cell death in Alzheimer's disease.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , tau Proteins/metabolism , tau Proteins/toxicity , Amyloid beta-Protein Precursor/pharmacology , Blotting, Western , Caspase 3 , Caspase Inhibitors , Cell Line , Cells, Cultured , Enzyme Inhibitors/pharmacology , Humans , Indicators and Reagents , Peptide Fragments/genetics , Peptide Fragments/toxicity , Plasmids/genetics , Recombinant Proteins/pharmacology , Transfection , beta-Galactosidase/biosynthesis , tau Proteins/genetics
10.
Brain Res ; 883(1): 60-8, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-11063988

ABSTRACT

NF-kappaB is a transcription factor, which is activated by various stimuli. One of the well-known activators of NF-kappaB is oxidative stress, which is a cause of cell death in some tissue, or cell types. Optic nerve transection, axotomy, results in retinal cell death, because of oxidative stress, deprivation of neurotrophic factors, etc. Since it has been hypothesized that the retinal ganglion cell death after axotomy is due to the generation of reactive oxygen species, we investigated whether NF-kappaB is involved in the retinal cell death after axotomy. This study was performed to investigate the role of NF-kappaB in retinal ganglion cell death after optic nerve transection. We used double staining experiment by using anti-NF-kappaB antibody and ethidium bromide to observe the correlation of NF-kappaB activation and the cell death. NF-kappaB was observed only in the surviving cells. NF-kappaB translocation was observed 3 days after the optic nerve transection. The NF-kappaB inhibitor, sulfasalazine, was used to block the activation of NF-kappaB in the axotomized retina, and the number of ganglion cells was quantified using retrograde in the presence or absence of sulfasalazine after axotomy. Inhibition of NF-kappaB by sulfasalazine accelerated the degeneration of ganglion cells in the retina. The results suggest that the activated NF-kappaB plays a protective role from the cell death in the injured ganglion cells.


Subject(s)
Apoptosis/physiology , NF-kappa B/physiology , Optic Nerve/physiology , Retinal Ganglion Cells/physiology , Animals , Axotomy , Cell Survival , Male , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/metabolism , Sulfasalazine/pharmacology
11.
J Neurochem ; 75(6): 2327-34, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11080184

ABSTRACT

We examined patterns and mechanisms of cell death induced by haloperidol. Cortical cell cultures exposed to 10-100 microM: haloperidol for 24 h underwent neuronal death without injuring glia. The degenerating neurons showed hallmarks of apoptosis, featuring cell body shrinkage, nuclear chromatin condensation and aggregation, nuclear membrane disintegration with intact plasma membrane, and prominent internucleosomal DNA fragmentation. Neither glutamate antagonists nor antioxidants prevented the haloperidol-induced neuronal apoptosis. The c-Jun-NH(2)-terminal protein kinase and p38 mitogen-activated protein kinase were activated within 1 h and were sustained over the next 3 h following exposure of cortical neurons to 30 microM haloperidol. Haloperidol-induced neuronal apoptosis was partially attenuated by 10-30 microM PD169316, a selective inhibitor of p38 mitogen-activated protein kinase. Inclusion of 1 microg/ml cycloheximide, a protein synthesis inhibitor, or 100 ng/ml insulin prevented activation of both kinases and subsequent neuronal death. The present study demonstrates that cortical neurons exposed to haloperidol undergo apoptosis depending on activation of p38 mitogen-activated protein kinase and c-Jun-NH(2)-terminal protein kinase sensitive to cycloheximide and insulin.


Subject(s)
Apoptosis , Haloperidol/toxicity , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cells, Cultured , Cycloheximide/pharmacology , DNA Fragmentation/drug effects , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Free Radicals/metabolism , Imidazoles/pharmacology , Insulin/pharmacology , JNK Mitogen-Activated Protein Kinases , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neurons/cytology , Neurons/enzymology , Protein Synthesis Inhibitors/pharmacology , p38 Mitogen-Activated Protein Kinases
12.
Neurobiol Dis ; 7(4): 251-9, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10964597

ABSTRACT

Neurotrophins render neurons highly vulnerable to certain injuries. We examined the possibility that NT-4/5 would enhance free radical neurotoxicity in vivo as well as in vitro. Striatal neurons exposed to 10 microM Fe(2+) or 1 mM l-buthionine-[S, R]-sulfoximine (BSO) underwent mild degeneration within 24 h. With concurrent addition of 10-100 ng/ml NT-4/5, neuronal death following exposure to Fe(2+) or BSO was significantly increased and suppressed by addition of 100 microM trolox, an antioxidant. In the adult brain, the intrastriatal injections of 20 nmol Fe(2+) revealed features of neuronal necrosis such as swelling cell body and mitochondria, fenestration of plasma membrane prior to nuclear membrane, and scattering condensation of nuclear chromatin. Cotreatment with 1.8 microg NT-4/5 augmented the striatal damage 24 h following the injections of Fe(2+). This study implies that free radicals produce necrotic degeneration in vivo as well as in vitro that becomes more sensitive in the presence of neurotrophins.


Subject(s)
Cell Death/drug effects , Nerve Growth Factors/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Cell Death/physiology , Cells, Cultured , Corpus Striatum , Embryo, Mammalian , Free Radicals/pharmacology , Necrosis , Nerve Degeneration/chemically induced , Neurons/pathology , Rats
13.
Article in English | MEDLINE | ID: mdl-10936764

ABSTRACT

Cellular and biochemical responses of the pectoral muscle to variation in seasonal activity were studied in the bat, Murina leucogaster ognevi. We collected bats in mid-hibernation (February), end-hibernation (April), and mid-summer (August) to track major activity periods in their annual cycle. Our findings indicated that myofiber cross-sectional area decreased to 68% between mid- and end-hibernation, but returned to the winter level in mid-summer. Total soluble protein and total RNA concentrations were not altered over these sampling periods. Oxidative potential gauged by citrate synthase activity increased 1.47-fold from mid- to end-hibernation and then remained at the similar level in mid-summer. Glycolytic potential gauged by lactate dehydrogenase activity changed little between mid- and end-hibernation but increased 1.42-fold in summer, compared with the winter level. Thus, the myofibers underwent disuse atrophy during hibernation, while enzymatic catalytic function recovered towards the level of mid-summer.


Subject(s)
Chiroptera/physiology , Muscle, Skeletal/physiology , Seasons , Adenosine Triphosphatases/metabolism , Animals , Dihydrolipoamide Dehydrogenase/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Muscle Proteins/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism
14.
J Neurochem ; 74(6): 2455-61, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10820206

ABSTRACT

We examined the possibility that p38 mitogen-activated protein kinase and caspase-3 would be activated for execution of apoptosis and excitotoxicity, the two major types of neuronal death underlying hypoxicischemic and neurodegenerative diseases. Mouse cortical cell cultures underwent widespread neuronal apoptosis 24 h following exposure to 10-30 nM calyculin A, a selective inhibitor of Ser/Thr phosphatase I and IIA. Activity of p38 was increased 2-4 h following exposure to 30 nM calyculin A. Addition of 3-10 microM PD169316, a selective p38 inhibitor, partially attenuated calyculin A neurotoxicity. Activity of caspase-3-like proteases was increased in cortical cell cultures exposed to 30 nM calyculin A for 8-16 h as shown by cleavage of DEVD-p-nitroanilide and phosphorylated tau. Proteolysis of tau was completely blocked by addition of 100 microM N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone (z-VAD-fmk), a broad-spectrum inhibitor of caspases, but incompletely by 10 microM PD169316. Calyculin A neurotoxicity was partially sensitive to 100 microM z-VAD-fmk. Cotreatment with 10 microM PD169316 and 100 microM z-VAD-fmk showed additive neuroprotection against calyculin A. Neither PD169316 nor z-VAD-fmk showed a beneficial effect against excitotoxic neuronal necrosis induced by exposure to 20 microM NMDA. Thus, caspase-3-like proteases and p38 likely contribute to calyculin A-induced neuronal apoptosis but not NMDA-induced neuronal necrosis.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Excitatory Amino Acid Agonists/pharmacology , Mitogen-Activated Protein Kinases/metabolism , N-Methylaspartate/pharmacology , Neurons/enzymology , Oxazoles/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Caspase 3 , Cells, Cultured , Cerebral Cortex/cytology , Cysteine Proteinase Inhibitors/pharmacology , Drug Synergism , Enzyme Inhibitors/pharmacology , Fetus/cytology , Imidazoles/pharmacology , Marine Toxins , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Necrosis , Neurons/chemistry , Neurons/pathology , Neurotoxins/pharmacology , Receptors, N-Methyl-D-Aspartate/physiology , p38 Mitogen-Activated Protein Kinases , tau Proteins/metabolism
15.
Neuroscience ; 94(3): 917-27, 1999.
Article in English | MEDLINE | ID: mdl-10579584

ABSTRACT

Consistent with previous studies on cell lines and non-neuronal cells, specific inhibitors of protein kinase C induced mouse primary cultured neocortical neurons to undergo apoptosis. To examine the complementary hypothesis that activating protein kinase C would attenuate neuronal apoptosis, the cultures were exposed for 1 h to phorbol-12-myristate-13-acetate, which activated protein kinase C as evidenced by downstream enhancement of the mitogen-activated protein kinase pathway. Exposure to phorbol-12-myristate-13-acetate, or another active phorbol ester, phorbol-12,13-didecanoate, but not to the inactive ester, 4alpha-phorbol-12,13-didecanoate, markedly attenuated neuronal apoptosis induced by serum deprivation. Phorbol-12-myristate-13-acetate also attenuated neuronal apoptosis induced by exposure to beta-amyloid peptide 1-42, or oxygen-glucose deprivation in the presence of glutamate receptor antagonists. The neuroprotective effects of phorbol-12-myristate-13-acetate were blocked by brief (non-toxic) concurrent exposure to the specific protein kinase C inhibitors, but not by a specific mitogen-activated protein kinase 1 inhibitor. Phorbol-12-myristate-13-acetate blocked the induction of p38 mitogen-activated protein kinase activity and specific inhibition of this kinase by SB 203580 attenuated serum deprivation-induced apoptosis. c-Jun N-terminal kinase 1 activity was high at rest and not modified by phorbol-12-myristate-13-acetate treatment. These data strengthen the idea that protein kinase C is a key modulator of several forms of central neuronal apoptosis, in part acting through inhibition of p38 mitogen-activated protein kinase regulated pathways.


Subject(s)
Cerebral Cortex/cytology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neurons/cytology , Neurons/physiology , Phorbol Esters/pharmacology , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Amyloid beta-Peptides/pharmacology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cell Hypoxia , Cells, Cultured , Coculture Techniques , Culture Media, Serum-Free , Cycloheximide/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Imidazoles/pharmacology , Indoles/pharmacology , JNK Mitogen-Activated Protein Kinases , Kinetics , Maleimides/pharmacology , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Neurons/drug effects , Peptide Fragments/pharmacology , Pyridines/pharmacology , Staurosporine/pharmacology , p38 Mitogen-Activated Protein Kinases
16.
Res Commun Mol Pathol Pharmacol ; 104(1): 31-41, 1999.
Article in English | MEDLINE | ID: mdl-10604276

ABSTRACT

Both direct and indirect environmental stress to the brain can increase the expression of transcription factor c-fos in various populations of neurons. In this study, we examined whether the intraperitoneal injections of lidocaine at doses inducing convulsions within 10 min, increased the level of c-fos mRNA and protein in forebrain areas. In in situ hybridization using [35S]UTP-labeled antisense c-fos, cRNA increased c-fos mRNA levels through the hippocampal formation, piriform cortex, septum, caudate-putamen, neostriatum, and amygdala within 2 hr. In parallel with the mRNA expression, c-fos protein immuno-reactivity was also observed in the same forebrain areas. In contrast to the seizure activity and wide-spread neuronal degeneration following kainate treatment, injections of lidocaine did not produce neuronal death within three days. The present study indicates that lidocaine induces convulsions and c-fos expression without causing neurotoxicity.


Subject(s)
Brain/drug effects , Gene Expression Regulation/drug effects , Lidocaine/pharmacology , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/genetics , Animals , Brain/metabolism , In Situ Hybridization , Injections, Intraperitoneal , Lidocaine/administration & dosage , Male , Rats , Rats, Sprague-Dawley , Seizures/chemically induced
17.
Neuroscience ; 94(1): 83-91, 1999.
Article in English | MEDLINE | ID: mdl-10613499

ABSTRACT

Administration of the excitotoxin kainate produces seizure activity and selective neuronal death in various brain areas. We examined the degeneration pattern of hippocampal neurons following systemic injections of kainate in the hamster and the rat. As reported, treatment with kainate resulted in severe neuronal loss in the hilus and CA3 in the rat. While the hilar neurons were also highly vulnerable to kainate in the hamster, neurons in the CA1 area, but not CA3, were highly sensitive to kainate. In both animals, immunoreactivity to anti-p50 nuclear factor kappa B antibody was increased in nuclei of the hilar neurons within 4 h following administration of kainate. Kainate treatment also increased the nuclear factor kappa B immunoreactivity in hamster CA1 neurons and rat CA3 neurons 24 h later. Neurons showing intense nuclear factor kappa B signal were stained with acid fuchsin. Kainate also increased DNA binding activity of p50 and p65 nuclear factor kappa B in the nuclear extract of the hippocampal formation as analysed by electrophoretic mobility shift assay in the hamster, suggesting that activation of nuclear factor kappa B may contribute to kainate-induced hippocampal degeneration. Administration of 100 nmol dizocilpine maleate 3 h prior to kainate attenuated kainate-induced activation of nuclear factor kappa B and neuronal death in CA1 in the hamster. The present study provides evidence that the differential vulnerability of neurons in the rat and the hamster hippocampus to kainate is partly mediated by mechanisms involving N-methyl-D-aspartate-dependent activation of nuclear factor kappa B.


Subject(s)
Excitatory Amino Acid Agonists/toxicity , Hippocampus/drug effects , Kainic Acid/toxicity , NF-kappa B/metabolism , Animals , Cell Death/drug effects , Cell Nucleus/metabolism , Cricetinae , Dizocilpine Maleate/pharmacology , Epilepsy/chemically induced , Epilepsy/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Female , Hippocampus/cytology , Hippocampus/metabolism , Mesocricetus , N-Methylaspartate/pharmacology , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Neurons/cytology , Neurons/drug effects , Neurotoxins/toxicity , Rats , Rats, Sprague-Dawley
18.
J Neurosci ; 19(20): 8849-55, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10516304

ABSTRACT

Cultured cortical neurons maintained in 25 mM glucose underwent a widespread neuronal death after exposure to NMDA, AMPA, and kainate. Among these, NMDA toxicity was substantially reduced in neurons maintained in 100 mM glucose. NMDA-induced increase in [Ca(2+)](i) and reactive oxygen species was attenuated in neurons maintained in high glucose that revealed increased mitochondrial membrane and redox potentials as determined using rhodamine 123 and 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide. p-trifluoromethoxy-phenylhydrazone, KCN, and rotenone, the selective inhibitors of mitochondrial potential, abrogated neuroprotective effect of high glucose against NMDA. The neuroprotective action of high glucose was extended against oxygen or combined oxygen-glucose deprivation. The present study provides evidence that prolonged exposure of cortical cells to high glucose attenuates NMDA- and free radical-mediated neuronal death via enhanced mitochondrial function.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Glucose/administration & dosage , Glucose/deficiency , Hypoxia/physiopathology , Mitochondria/physiology , N-Methylaspartate/pharmacology , Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Electrophysiology , Glucose/pharmacology , Hypoxia/pathology , Mice , Mice, Inbred ICR , Neurons/drug effects , Neurons/physiology , Neurotoxins/metabolism
19.
Exp Neurol ; 159(1): 217-24, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10486189

ABSTRACT

We examined the possibility that catecholamines (CA) could act as endogenous modulators of neuronal death. Exposure to high doses (>100 microM) of dopamine (DA) caused widespread neuronal death within 24 h in mouse cortical cell cultures and was accompanied by cell body shrinkage, aggregation and condensation of nuclear chromatin, and prominent internucleosomal DNA fragmentation. Epinephrine, but not norepinephrine (NE), was slightly toxic to neurons at doses higher than 1 mM. DA-induced death was attenuated by the addition of three different anti-apoptosis agents, 1 microgram/ml cycloheximide, 25 mM K(+), or 100 ng/ml brain-derived neurotrophic factor (BDNF). While treatment with 100 microM N-acetyl-l-cysteine attenuated DA neurotoxicity, neither the glutamate antagonists (10 microM MK-801 plus 50 microM CNQX) nor several antioxidants [trolox, 100 microM; Mn (III) tetrakis (4-benzoic acid) porphyrin chloride, 100 microM; Mn (III) tetrakis (1-methyl-4-pyridyl) prophyrin pentachloride, 100 microM; N-tert-butyl-alpha-phenylnitrone, 3 mM] prevented the CA-induced apoptosis. Interestingly, all CA at 1-30 microM attenuated free radical-mediated neuronal necrosis following exposure to 30 microM Fe(2+) or 200 microM H(2)O(2), which was insensitive to DA or NE antagonists. Like trolox, CA reduced levels of the stable free radical 1,1-diphenyl-2-picrylhydrazyl under cell-free conditions, raising the possibility that CA as an antioxidant protects neurons. We also found that the neuroprotective effect of CA prolonged the protective effects of BDNF against serum deprivation. The present findings suggest that CA induces apoptosis at high doses but prevents free radical-mediated neurotoxicity as an anti-oxidant without being coupled to the receptors.


Subject(s)
Apoptosis/drug effects , Catecholamines/toxicity , Neurons/cytology , Acetylcysteine/metabolism , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cardiotonic Agents/toxicity , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cycloheximide/pharmacology , DNA Damage , Dopamine/toxicity , Dose-Response Relationship, Drug , Drug Interactions , Epinephrine/toxicity , Fetus/cytology , Free Radicals/metabolism , Lipid Peroxidation , Mice , Neurons/drug effects , Neurons/metabolism , Norepinephrine/toxicity , Potassium/pharmacology , Protein Synthesis Inhibitors/pharmacology , Sympathomimetics/toxicity
20.
Exp Neurol ; 159(1): 274-82, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10486196

ABSTRACT

To further characterize MPP(+)-induced cell death and to explore the role of Bcl-2-related proteins in this death paradigm, we utilized a mesencephalon-derived dopaminergic neuronal cell line (MN9D) stably transfected with human bcl-2 (MN9D/Bcl-2), its C-terminal deletion mutant (MN9D/Bcl-2Delta22), murine bax (MN9D/Bax), or a control vector (MN9D/Neo). As determined by electron microscopy and TUNEL assay, MN9D/Neo cells exposed to MPP(+) underwent a cell death that was characterized by mitochondrial swelling and irregularly scattered heterochromatin without accompanying DNA fragmentation. However, cell swelling typically seen in necrosis did not appear. To examine the biochemical events associated with MPP(+)-induced cell death, various analyses were conducted. Addition of a broad-spectrum caspase inhibitor, N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (50-400 microM) or Boc-aspartyl(OMe)-fluoromethylketone (50-200 microM) did not attenuate MPP(+)-induced cell death while the same treatment protected MN9D/Neo cells against staurosporine-induced apoptotic cell death. Concurrent treatment with an inhibitor of macromolecule synthesis such as cycloheximide, emetine, or actinomycin D blocked MPP(+)-induced cell death, suggesting that new protein synthesis is required as demonstrated in many apoptotic cell death. The level of cytosolic calcium in MN9D/Neo cells was unchanged over 24 h following MPP(+) treatment, as monitored by means of the fluorescent probe Fura-2. Western blot analysis indicated that expression level of proapoptotic protein, Bax was not significantly altered after MPP(+) treatment. In this death paradigm, overexpression of Bcl-2 but not its C-terminal deletion mutant attenuated MPP(+)-induced cell death whereas overexpression of Bax had no effect. Taken together, these data indicate that (i) MPP(+) induces a distinct form of cell death which resembles both apoptosis and necrosis; and (ii) full-length Bcl-2 counters MPP(+)-induced morphological changes and cell death via a mechanism that is dependent on de novo protein synthesis but independent of cytosolic calcium changes, Bax expression, and/or activation of caspase(s) in MN9D cells.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Calcium/metabolism , Caspases/metabolism , Cell Death/drug effects , Dopamine Agents/toxicity , Neurons/cytology , Proto-Oncogene Proteins c-bcl-2/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Cell Line , Cycloheximide/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Cytosol/metabolism , Dactinomycin/pharmacology , Dopamine/physiology , Emetine/pharmacology , Enzyme Inhibitors/pharmacology , Humans , In Situ Nick-End Labeling , Mesencephalon/cytology , Mice , Microscopy, Electron , Nerve Tissue Proteins/biosynthesis , Neurons/enzymology , Neurons/ultrastructure , Neuroprotective Agents/pharmacology , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins/metabolism , Staurosporine/pharmacology , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...