Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 6: e1645, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25675305

ABSTRACT

The glial cell line-derived neurotrophic factor (GDNF) has an important role in neuronal survival through binding to the GFRα1 (GDNF family receptor alpha-1) receptor and activation of the receptor tyrosine kinase Ret. Transient brain ischemia alters the expression of the GDNF signaling machinery but whether the GDNF receptor proteins are also affected, and the functional consequences, have not been investigated. We found that excitotoxic stimulation of cultured hippocampal neurons leads to a calpain-dependent downregulation of the long isoform of Ret (Ret51), but no changes were observed for Ret9 or GFRα1 under the same conditions. Cleavage of Ret51 by calpains was selectively mediated by activation of the extrasynaptic pool of N-methyl-d-aspartate receptors and leads to the formation of a stable cleavage product. Calpain-mediated cleavage of Ret51 was also observed in hippocampal neurons subjected to transient oxygen and glucose deprivation (OGD), a model of global brain ischemia, as well as in the ischemic region in the cerebral cortex of mice exposed to transient middle cerebral artery occlusion. Although the reduction of Ret51 protein levels decreased the total GDNF-induced receptor activity (as determined by assessing total phospho-Ret51 protein levels) and their downstream signaling activity, the remaining receptors still showed an increase in phosphorylation after incubation of hippocampal neurons with GDNF. Furthermore, GDNF protected hippocampal neurons when present before, during or after OGD, and the effects under the latter conditions were more significant in neurons transfected with human Ret51. These results indicate that the loss of Ret51 in brain ischemia partially impairs the neuroprotective effects of GDNF.


Subject(s)
Brain Ischemia/metabolism , Calpain/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hippocampus/cytology , Neurons/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Animals , Cells, Cultured , Glutamic Acid/pharmacology , Humans , Mice , Neurons/cytology , Rats , Signal Transduction/drug effects
2.
Cell Death Dis ; 4: e612, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23640454

ABSTRACT

Recent studies have shown that type 2 diabetes mellitus (T2DM) is a risk factor for cognitive dysfunction or dementia. Insulin resistance is often associated with T2DM and can induce defective insulin signaling in the central nervous system as well as increase the risk of cognitive impairment in the elderly. Glucagone like peptide-1 (GLP-1) is an incretin hormone and, like GLP-1 analogs, stimulates insulin secretion and has been employed in the treatment of T2DM. GLP-1 and GLP-1 analogs also enhance synaptic plasticity and counteract cognitive deficits in mouse models of neuronal dysfunction and/or degeneration. In this study, we investigated the potential neuroprotective effects of long-term treatment with exenatide, a GLP-1 analog, in two animal models of neuronal dysfunction: the PS1-KI and 3xTg-AD mice. We found that exenatide promoted beneficial effects on short- and long-term memory performances in PS1-KI but not in 3xTg-AD animals. In PS1-KI mice, the drug increased brain lactate dehydrogenase activity leading to a net increase in lactate levels, while no effects were observed on mitochondrial respiration. On the contrary, exenatide had no effects on brain metabolism of 3xTg-AD mice. In summary, our data indicate that exenatide improves cognition in PS1-KI mice, an effect likely driven by increasing the brain anaerobic glycolysis rate.


Subject(s)
Brain/drug effects , Hypoglycemic Agents/pharmacology , Peptides/pharmacology , Venoms/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Brain/enzymology , Brain/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Cognition Disorders/pathology , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Electron Transport Complex IV/metabolism , Exenatide , Female , Hypoglycemic Agents/therapeutic use , L-Lactate Dehydrogenase/metabolism , Lactic Acid/metabolism , Male , Memory, Long-Term/drug effects , Memory, Short-Term/drug effects , Mice , Mice, Transgenic , Mitochondria/enzymology , Peptides/therapeutic use , Venoms/therapeutic use , tau Proteins/metabolism
3.
Cell Death Dis ; 3: e448, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-23254291

ABSTRACT

In this study, we investigated the effects of long-term (9-month) treatment with pioglitazone (PIO; 20 mg/kg/d) in two animal models of Alzheimer's disease (AD)-related neural dysfunction and pathology: the PS1-KI(M146V) (human presenilin-1 (M146V) knock-in mouse) and 3xTg-AD (triple transgenic mouse carrying AD-linked mutations) mice. We also investigated the effects on wild-type (WT) mice. Mice were monitored for body mass changes, fasting glycemia, glucose tolerance, and studied for changes in brain mitochondrial enzyme activity (complexes I and IV) as well as energy metabolism (lactate dehydrogenase (LDH)). Cognitive effects were investigated with the Morris water maze (MWM) test and the object recognition task (ORT). Behavioral analysis revealed that PIO treatment promoted positive cognitive effects in PS1-KI female mice. These effects were associated with normalization of peripheral gluco-regulatory abnormalities that were found in untreated PS1-KI females. PIO-treated PS1-KI females also showed no statistically significant alterations in brain mitochondrial enzyme activity but significantly increased reverse LDH activity.PIO treatment produced no effects on cognition, glucose metabolism, or mitochondrial functioning in 3xTg-AD mice. Finally, PIO treatment promoted enhanced short-term memory performance in WT male mice, a group that did not show deregulation of glucose metabolism but that showed decreased activity of complex I in hippocampal and cortical mitochondria. Overall, these results indicate metabolically driven cognitive-enhancing effects of PIO that are differentially gender-related among specific genotypes.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Cognition/drug effects , Glucose/metabolism , Presenilin-1/genetics , Thiazolidinediones/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/psychology , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Pioglitazone , Presenilin-1/metabolism , Time Factors
4.
Cell Death Dis ; 1: e91, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-21368864

ABSTRACT

The overall effect of brain zinc (Zn(2+)) in the progression and development of Alzheimer's disease (AD) is still not completely understood. Although an excess of Zn(2+) can exacerbate the pathological features of AD, a deficit of Zn(2+) intake has also been shown to increase the volume of amyloid plaques in AD transgenic mice. In this study, we investigated the effect of dietary Zn(2+) supplementation (30 p.p.m.) in a transgenic mouse model of AD, the 3xTg-AD, that expresses both ß amyloid (Aß)- and tau-dependent pathology. We found that Zn(2+) supplementation greatly delays hippocampal-dependent memory deficits and strongly reduces both Aß and tau pathology in the hippocampus. We also evaluated signs of mitochondrial dysfunction and found that Zn(2+) supplementation prevents the age-dependent respiratory deficits we observed in untreated 3xTg-AD mice. Finally, we found that Zn(2+) supplementation greatly increases the levels of brain-derived neurotrophic factor (BDNF) of treated 3xTg-AD mice. In summary, our data support the idea that controlling the brain Zn(2+) homeostasis may be beneficial in the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Brain-Derived Neurotrophic Factor/metabolism , Cognition Disorders/prevention & control , Mitochondria/physiology , Zinc/therapeutic use , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Dietary Supplements , Hippocampus/pathology , Mice , Mice, Transgenic , Zinc/administration & dosage , tau Proteins/metabolism
5.
Cell Death Dis ; 1: e90, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-21368863

ABSTRACT

The triple-transgenic Alzheimer (3 × Tg-AD) mouse expresses mutant PS1(M146V), APP(swe), and tau(P301L) transgenes and progressively develops plaques and neurofibrillary tangles with a temporal- and region-specific profile that resembles the neuropathological progression of Alzheimer's disease (AD). In this study, we used proteomic approaches such as two-dimensional gel electrophoresis and mass spectrometry to investigate the alterations in protein expression occurring in the brain and cerebellum of 3 × Tg-AD and presenilin-1 (PS1) knock-in mice (animals that do not develop Aß- or tau-dependent pathology nor cognitive decline and were used as control). Finally, using the Ingenuity Pathway Analysis we evaluated novel networks and molecular pathways involved in this AD model. We identified several differentially expressed spots and analysis of 3 × Tg-AD brains showed a significant downregulation of synaptic proteins that are involved in neurotransmitter synthesis, storage and release, as well as a set of proteins that are associated with cytoskeleton assembly and energy metabolism. Interestingly, in the cerebellum, a structure not affected by AD, we found an upregulation of proteins involved in carbohydrate metabolism and protein catabolism. Our findings help to unravel the pathogenic brain mechanisms set in motion by mutant amyloid precursor protein (APP) and hyperphosphorylated tau. These data also reveal cerebellar pathways that may be important to counteract the pathogenic actions of Aß and tau, and ultimately offer novel targets for therapeutic intervention.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Brain/metabolism , Cerebellum/metabolism , Proteome/metabolism , tau Proteins/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional , Female , Gene Knock-In Techniques , Mice , Mice, Transgenic , Presenilin-1/genetics , Presenilin-1/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , tau Proteins/metabolism
6.
J Neurophysiol ; 98(4): 2274-84, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17699699

ABSTRACT

We employed whole cell patch-clamp recordings to establish the effect of Zn(2+) on the gating the brain specific, T-type channel isoform Ca(V)3.3 expressed in HEK-293 cells. Zn(2+) (300 microM) modified the gating kinetics of this channel without influencing its steady-state properties. When inward Ca(2+) currents were elicited by step depolarizations at voltages above the threshold for channel opening, current inactivation was significantly slowed down while current activation was moderately affected. In addition, Zn(2+) slowed down channel deactivation but channel recovery from inactivation was only modestly changed. Zn(2+) also decreased whole cell Ca(2+) permeability to 45% of control values. In the presence of Zn(2+), Ca(2+) currents evoked by mock action potentials were more persistent than in its absence. Furthermore, computer simulation of action potential generation in thalamic reticular cells performed to model the gating effect of Zn(2+) on T-type channels (while leaving the kinetic parameters of voltage-gated Na(+) and K(+) unchanged) revealed that Zn(2+) increased the frequency and the duration of burst firing, which is known to depend on T-type channel activity. In line with this finding, we discovered that chelation of endogenous Zn(2+) decreased the frequency of occurrence of ictal-like epileptiform discharges in rat thalamocortical slices perfused with medium containing the convulsant 4-aminopyridine (50 microM). These data demonstrate that Zn(2+) modulates Ca(V)3.3 channel gating thus leading to increased neuronal excitability. We also propose that endogenous Zn(2+) may have a role in controlling thalamocortical oscillations.


Subject(s)
Calcium Channels, T-Type/drug effects , Cerebral Cortex/physiology , Ion Channel Gating/drug effects , Membrane Transport Proteins/drug effects , Thalamus/physiology , Zinc/pharmacology , 4-Aminopyridine/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Algorithms , Animals , Calcium Channels, T-Type/genetics , Cell Line , Cerebral Cortex/drug effects , Chelating Agents/pharmacology , Data Interpretation, Statistical , Epilepsy/chemically induced , Epilepsy/physiopathology , Humans , In Vitro Techniques , Kinetics , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Transport Proteins/genetics , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Thalamus/drug effects , Transfection
7.
J Neurochem ; 103(4): 1472-80, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17680990

ABSTRACT

To identify the transductional mechanisms responsible for the neuroprotective effect of nitric oxide (NO) during ischemic preconditioning (IPC), we investigated the effects of this gaseous mediator on mitochondrial Mn-superoxide dismutase (Mn-SOD) expression and activity. In addition, the possible involvement of Ras/extracellular-regulated kinase (ERK) ERK1/2 pathway in preserving cortical neurons exposed to oxygen and glucose deprivation (OGD) followed by reoxygenation was also examined. Ischemic preconditioning was obtained by exposing neurons to a 30-min sublethal OGD (95% N(2) and 5% CO(2)). Then, after a 24-h interval, neurons were exposed to 3 h of OGD followed by 24 h of reoxygenation (OGD/Rx). Our results revealed that IPC reduced cytochrome c (cyt c) release into the cytosol, improved mitochondrial function, and decreased free radical production. Moreover, it induced an increase in nNOS expression and NO production and promoted ERK1/2 activation. These effects were paralleled by an increase in Mn-SOD expression and activity that persisted throughout the following OGD phase. When the neurons were treated with L-NAME, a well known NOS inhibitor, the increase in Mn-SOD expression occurring during IPC was reduced and, as a result, IPC-induced neuroprotection was prevented. Similarly, when ERK1/2 was inhibited by its selective inhibitor PD98059, the increase in Mn-SOD expression observed during IPC was almost completely abolished. As a result, its neuroprotective effect on cellular survival was thwarted. The present findings indicate that during IPC the increase in Mn-SOD expression and activity are paralleled by NO production. This suggests that NO neuroprotective role occurs through the stimulation of Mn-SOD expression and activity. In particular, NO via Ras activation stimulates downstream ERK1/2 cascade. This pathway, in turn, post-transcriptionally activates Mn-SOD expression and activity, thus promoting neuroprotection during preconditioning.


Subject(s)
Ischemic Preconditioning , MAP Kinase Signaling System/physiology , Mitochondrial Proteins/metabolism , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Nitric Oxide/physiology , Superoxide Dismutase/metabolism , ras Proteins/physiology , Animals , Cell Survival/physiology , Cells, Cultured , Enzyme Activation/physiology , Gene Expression Regulation/physiology , Ischemic Preconditioning/methods , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Neuroprotective Agents/metabolism , Rats , Rats, Wistar , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics
8.
J Neurosci Res ; 76(6): 812-21, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15160393

ABSTRACT

The present study investigated the temporal relationship between neuronal nitric oxide synthase (nNOS) activity and expression and the development of neuronal damage occurring during anoxia and anoxia followed by reoxygenation. For this purpose, cerebellar granule cells were exposed to 2 hr of oxygen and glucose deprivation (OGD) and 24 hr of reoxygenation. To clarify the consequences of nNOS activity inhibition on neuronal survival, cerebellar granule cells were exposed to OGD, both in the absence of extracellular Na(+) ([Na(+)](e)), a condition that by reducing intracellular Ca(2+) ([Ca(2+)](I)) prevents Ca(2+)-dependent nNOS activation, and in the presence of selective and nonselective nNOS inhibitors, such as N(omega)-L-allyl-L-arginine (L-ALA), N(omega)-propyl-L-arginine (NPLA), and L-nitro-arginine-methyl-ester (L-NAME), respectively. The results demonstrated that the removal of [Na(+)](e) hampered the [Ca(2+)](i) increase and decreased expression and activity of nNOS. Similarly, the increase of free radical production present in cerebellar neurons, exposed previously to OGD and OGD/reoxygenation, was abolished completely in the absence of [Na(+)](e). Furthermore, the absence of [Na(+)](e) in cerebellar neurons exposed to 2 hr of OGD led to the improvement of mitochondrial activity and neuronal survival, both after the OGD phase and after 24 hr of reoxygenation. Finally, the exposure of cerebellar neurons to L-ALA (200 nM), and L-NAME (500 microM) was able to effectively reduce NO(*) production and caused an increase in mitochondrial oxidative activity and an improvement of neuronal survival not only during OGD, but also during reoxygenation. Similar results during OGD were obtained also with NPLA (5 nM), another selective nNOS inhibitor. These data suggest that the activation of nNOS is highly accountable for the neuronal damage occurring during the OGD and reoxygenation phases.


Subject(s)
Brain Ischemia/enzymology , Cerebellum/enzymology , Glucose/deficiency , Neurons/enzymology , Nitric Oxide Synthase/metabolism , Reperfusion Injury/enzymology , Animals , Brain Ischemia/physiopathology , Calcium/metabolism , Cell Death/physiology , Cell Hypoxia/physiology , Cells, Cultured , Cerebellum/cytology , Cerebellum/physiopathology , Enzyme Activation , Glucose/metabolism , Hypoxia/enzymology , Hypoxia/physiopathology , L-Lactate Dehydrogenase/metabolism , Neurons/pathology , Nitric Oxide Synthase Type I , Oxidative Stress/physiology , Oxygen/metabolism , Rats , Reperfusion Injury/physiopathology , Sodium/metabolism , Tetrazolium Salts/metabolism , Thiazoles/metabolism
9.
Proc Natl Acad Sci U S A ; 98(20): 11055-61, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11572968

ABSTRACT

Zinc is coreleased with glutamate from excitatory nerve terminals throughout the central nervous system and acutely inhibits N-methyl-d-aspartate (NMDA) receptor activation. Here we report that cultured murine cortical neurons briefly exposed to sublethal concentrations of zinc developed increased intracellular free Na(+), phosphorylation of Src kinase at tyrosine 220, and tyrosine phosphorylation of NMDA receptor 2A/2B subunits, in a fashion sensitive to the Src family kinase inhibitor 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine, PP2. Functionally, this zinc exposure produced a delayed increase in NMDA receptor current in perforated patch but not conventional whole-cell recordings, as well as an increase in NMDA receptor-mediated cell death. These observations suggest that the effect of synaptically released zinc on neuronal NMDA receptors may be biphasic: acute block, followed by Src family kinase-mediated up-regulation of NMDA receptor activity and cytotoxicity.


Subject(s)
Cerebral Cortex/physiology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Zinc/pharmacology , src-Family Kinases/metabolism , Animals , Cell Death , Cells, Cultured , Cerebral Cortex/cytology , Fetus , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , N-Methylaspartate/pharmacology , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques , Phosphorylation , Protein Subunits , Receptors, N-Methyl-D-Aspartate/drug effects , Sodium/metabolism , Tyrosine , Up-Regulation/drug effects
10.
Curr Opin Cell Biol ; 13(4): 405-11, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11454444

ABSTRACT

Alterations in the transmembrane gradients of several physiological ions may influence programmed cell death. In particular, recent data suggest that increases in intracellular calcium may either promote or inhibit apoptosis, depending on the level, timing and location, whereas loss of intracellular potassium promotes apoptosis.


Subject(s)
Apoptosis , Ion Transport , Animals , Calcium Signaling , Cell Size , Chloride Channels/physiology , Homeostasis , Models, Biological , Potassium/metabolism
11.
Neuroscience ; 100(3): 651-61, 2000.
Article in English | MEDLINE | ID: mdl-11098127

ABSTRACT

The translocation of synaptic Zn(2+) from nerve terminals into selectively vulnerable neurons may contribute to the death of these neurons after global ischemia. We hypothesized that cellular Zn(2+) overload might be lethal for reasons similar to cellular Ca(2+) overload and tested the hypothesis that Zn(2+) neurotoxicity might be mediated by the activation of nitric oxide synthase. Although Zn(2+) (30-300microM) altered nitric oxide synthase activity in cerebellar extracts in solution, it did not affect nitric oxide synthase activity in cultured murine neocortical neurons. Cultured neurons exposed to 300-500microM Zn(2+) for 5min under depolarizing conditions developed widespread degeneration over the next 24h that was unaffected by the concurrent addition of the nitric oxide synthase inhibitor N(G)-nitro-L-arginine. Furthermore, Zn(2+) neurotoxicity was attenuated when nitric oxide synthase activity in the cultures was induced by exposure to cytokines, exogenous nitric oxide was added or nitric oxide production was pharmacologically enhanced. The unexpected protective effect of nitric oxide against Zn(2+) toxicity may be explained, at least in part, by reduction of toxic Zn(2+) entry. Exposure to nitric oxide donors reduced Ba(2+) current through high-voltage activated calcium channels, as well as K(+)-stimulated neuronal uptake of 45Ca(2+) or 65Zn(2+). The oxidizing agents thimerosal and 2,2'-dithiodipyridine also reduced K(+)-stimulated cellular 45Ca(2+) uptake, while akylation of thiols by pretreatment with N-ethylmaleimide blocked the reduction of 45Ca(2+) uptake by a nitric oxide donor.The results suggest that Zn(2+)-induced neuronal death is not mediated by the activation of nitric oxide synthase; rather, available nitric oxide may attenuate Zn(2+) neurotoxicity by reducing Zn(2+) entry through voltage-gated Ca(2+) channels, perhaps by oxidizing key thiol groups.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Enzyme Inhibitors/pharmacology , Neurotoxins/pharmacology , Nitric Oxide/pharmacology , Zinc/metabolism , Zinc/pharmacology , Animals , Calcium Channels/drug effects , Electrophysiology , Mice , Neurons/enzymology , Neurotoxins/antagonists & inhibitors , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Oxidation-Reduction , Solutions , Zinc/antagonists & inhibitors
12.
J Physiol ; 528 Pt 1: 39-52, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11018104

ABSTRACT

1. Mammalian neuronal voltage-gated Ca2+ channels have been implicated as potential mediators of membrane permeability to Zn2+. We tested directly whether voltage-gated Ca2+ channels can flux Zn2+ in whole-cell voltage-clamp recordings from cultured murine cortical neurones. 2. In the presence of extracellular Zn2+ and no Na+, K+, or other divalent cations, a small, non-inactivating, voltage-gated inward current was observed exhibiting a current-voltage relationship characteristic of high-voltage activated (HVA) Ca2+ channels. Inward current was detectable at Zn2+ levels as low as 50 microM, and both the amplitude and voltage sensitivity of the current depended upon Zn2+ concentration. This Zn2+ current was sensitive to blockade by Gd3+ and nimodipine and, to a lesser extent, by omega-conotoxin GVIA. 3. Zn2+ could permeate Ca2+ channels in the presence of Ca2+ and other physiological cations. Inward currents recorded with 2 mM Ca2+ were attenuated by Zn2+ (IC50 = 210 microM), and currents recorded with Zn2+ were unaffected by up to equimolar Ca2+ concentrations. Furthermore, the Zn2+-selective fluorescent dye Newport Green revealed a depolarisation-activated, nimodipine-sensitive Zn2+ influx into cortical neurones that were bathed in a physiological extracellular solution plus 300 microM ZnCl2. 4. Surprisingly, while lowering extracellular pH suppressed HVA Ca2+ currents, Zn2+ current amplitude was affected oppositely, varying inversely with pH with an apparent pK of 7.4. The acidity-induced enhancement of Zn2+ current was associated with a positive shift in reversal potential but no change in the kinetics or voltage sensitivity of channel activation. 5. These results provide evidence that L- and N-type voltage-gated Ca2+ channels can mediate Zn2+ entry into cortical neurones and that this entry may be enhanced by extracellular acidity.


Subject(s)
Acids/metabolism , Calcium Channels/metabolism , Extracellular Space/metabolism , Neurons/metabolism , Zinc/metabolism , Acids/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cell Membrane Permeability/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Extracellular Space/drug effects , Fluorescent Dyes , Hydrogen-Ion Concentration/drug effects , Ion Transport/drug effects , Mice , Neurons/cytology , Patch-Clamp Techniques , Zinc/pharmacology
13.
Cell Mol Biol (Noisy-le-grand) ; 46(4): 797-806, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10875441

ABSTRACT

Although Zn2+ is normally stored and released in the brain, excessive exposure to extracellular Zn2+ can be neurotoxic. The purpose of the present study was to determine the type of neuronal cell death, necrosis versus apoptosis, induced by Zn2+ exposure. Addition of 10-50 microM ZnCl2 to the bathing medium of murine neuronal and glial cell cultures induced, over the next 24 hrs., Zn2+-concentration-dependent neuronal death; some glial death also occurred with Zn2+ concentrations above 30 microM. The neuronal death induced by 20 microM Zn2+ was characterized by coarse chromatin condensation, the formation of apoptotic bodies, and internucleosomal DNA fragmentation. It was attenuated in cortical cell cultures prepared from mice null for the bax gene, and by the caspase inhibitor, benzyloxycarbonyl-Val-Ala-Asp-CH2F (ZVAD, 100 microM), but not by the NMDA receptor antagonist, D-2-amino-5-phosphonovalerate (D-APV, 200 microM ). In contrast, the neuronal death induced by 50 microM Zn2+ was characterized by plasma membrane disruption and random DNA fragmentation; this death was attenuated by D-APV, but exhibited little sensitivity to ZVAD or deletion of bax. These results suggest that Zn2+ can induce cell death with characteristics of either apoptosis or necrosis, depending on the intensity of the Zn2+ exposure.


Subject(s)
Apoptosis/drug effects , Necrosis , Neurons/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-bcl-2 , Zinc/pharmacology , 2-Amino-5-phosphonovalerate/pharmacology , Alleles , Animals , Cell Membrane/drug effects , Chromatin/metabolism , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Electrophoresis, Agar Gel , Excitatory Amino Acid Antagonists/pharmacology , Genotype , L-Lactate Dehydrogenase/metabolism , Mice , Microscopy, Confocal , Microscopy, Electron , Neuroglia/drug effects , Neurons/cytology , Neurons/ultrastructure , Oligopeptides/pharmacology , Proto-Oncogene Proteins/genetics , bcl-2-Associated X Protein
14.
J Neurosci ; 19(19): RC31, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10493776

ABSTRACT

Toxic zinc influx may contribute to selective neuronal death after transient global ischemia. We previously used the high-affinity (K(D) = 27 nm) fluorescent dye mag-fura-5 to detect initial increases in neuronal intracellular free Zn(2+) ([Zn(2+)](i)) associated with brief Zn(2+) exposure. Here we used the specific low-affinity Zn(2+) indicator Newport Green (K(D) = 1 microm) to measure the peak levels of [Zn(2+)](i) attained during prolonged, toxic exposures to extracellular Zn(2+). Murine cortical cell cultures exposed for 5-10 min to 300 microm Zn(2+) in the presence of kainate or elevated extracellular K(+) developed widespread neuronal death over the next 24 hr. Such Zn(2+) exposure under depolarizing conditions was accompanied by a large increase in [Zn(2+)](i) reaching several hundred nanomolar, which gradually recovered over the next 20-40 min after termination of Zn(2+) exposure. Both the level of [Zn(2+)](i) elevation and the extent of subsequent neuronal death depended on the concentration of extracellular Zn(2+) between 30 microm and 1 mm. In contrast, exposure to 300 microm Zn(2+) in the presence of 300 microm NMDA resulted in little increase in [Zn(2+)](i) and little neuronal death, suggesting that NMDA receptor-gated channels are less important as a route of toxic Zn(2+) entry than voltage-gated calcium channels.


Subject(s)
Neurons/metabolism , Zinc/metabolism , Animals , Cell Death , Cells, Cultured , Excitatory Amino Acid Agonists/pharmacology , Extracellular Space/metabolism , Fetus , Fluorescent Dyes , Intracellular Fluid/metabolism , Kainic Acid/pharmacology , Mice , Microscopy, Confocal , N-Methylaspartate/pharmacology , Neocortex/cytology , Neocortex/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/drug effects , Potassium/pharmacology , Zinc/pharmacology
15.
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
16.
Neuroscience ; 90(4): 1339-48, 1999.
Article in English | MEDLINE | ID: mdl-10338301

ABSTRACT

Cultured cortical neurons exposed for 24 h to low concentrations of the Ca2+ ionophores, ionomycin (250 nM) or A-23187 (100 nM), underwent apoptosis, accompanied by early degeneration of neurites, cell body shrinkage, chromatin condensation and internucleosomal DNA fragmentation. This death could be blocked by protein synthesis inhibitors, as well as by the growth factors brain-derived neurotrophic factor or insulin-like growth factor I. If the ionomycin concentration was increased to 1-3 microM, then neurons underwent necrosis, accompanied by early cell body swelling without DNA laddering, or sensitivity to cycloheximide or growth factors. Calcium imaging with Fura-2 suggested a possible basis for the differential effects of low and high concentrations of ionomycin. At low concentrations, ionomycin induced greater increases in intracellular Ca2+ concentration in neurites than in neuronal cell bodies, whereas at high concentrations, ionomycin produced large increases in intracellular Ca2+ concentration in both neurites and cell bodies. We hypothesize that the ability of low concentrations of Ca2+ ionophores to raise intracellular Ca2+ concentration preferentially in neurites caused early neurite degeneration, leading to loss of growth factor availability to the cell body and consequent apoptosis, whereas high concentrations of ionophores produced global cellular Ca2+ overload and consequent necrosis.


Subject(s)
Apoptosis/physiology , Calcimycin/pharmacology , Calcium/metabolism , Ionomycin/pharmacology , Ionophores/pharmacology , Neocortex/cytology , Neurons/drug effects , Animals , Cells, Cultured , Mice , Necrosis , Neocortex/metabolism , Neocortex/pathology , Neurons/pathology , Neurons/physiology
17.
Eur J Neurosci ; 10(9): 2848-55, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9758154

ABSTRACT

Activation of ion channel-linked glutamate receptors, especially N-methyl-D-aspartate (NMDA) receptors, mediates the excitotoxic effects of glutamate upon central neurons. We examined the hypothesis that activation of group I metabotropic glutamate receptors (mGluRs) would increase NMDA receptor-mediated cortical neuronal death. Addition of the selective group I mGluR agonists, dihydroxyphenylglycine (DHPG) or trans-azetidine-2,4-dicarboxylic acid (t-ADA) potentiated NMDA-induced neuronal death, and application of the group I mGluR-selective antagonist, aminoindan-1,5-dicarboxylic acid (AIDA), as well as the non-selective antagonists methyl-4-carboxyphenylglycine (MCPG) or 4-carboxyphenylglycine (4CPG) reduced NMDA- and kainate-induced neuronal death in murine cortical cultures. The pro-excitotoxic effect of group I mGluR activation may be mediated largely by enhancement of glutamate release, as DHPG potentiated high potassium-stimulated glutamate release, and the protective effects of both AIDA and MCPG were abolished when NMDA and alpha-amino-3-hydroxy-5-methyl-4-isoxazole proprionic acid (AMPA) receptors were blocked immediately after toxic NMDA receptor overstimulation. The present data support the possibility that antagonizing group I mGluRs may be a useful strategy for attenuating excitotoxic neuronal death in certain disease states.


Subject(s)
Neurons/drug effects , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Animals , Astrocytes/cytology , Benzoates/pharmacology , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/analysis , Glutamic Acid/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Indans/pharmacology , Mice , N-Methylaspartate/pharmacology , Neurons/cytology , Neurotransmitter Agents/metabolism , Protein Binding/drug effects , Receptors, AMPA/drug effects , Receptors, Metabotropic Glutamate/biosynthesis , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects
18.
J Neurosci ; 18(16): 6290-9, 1998 Aug 15.
Article in English | MEDLINE | ID: mdl-9698321

ABSTRACT

The extracellular acidity that accompanies brain hypoxia-ischemia is known to reduce both NMDA and AMPA-kainate receptor-mediated currents and NMDA receptor-mediated neurotoxicity. Although a protective effect of acidic pH on AMPA-kainate receptor-mediated excitotoxicity has been assumed, such has not been demonstrated. Paradoxically, we found that lowering extracellular pH selectively increased AMPA-kainate receptor-mediated neurotoxicity in neocortical cell cultures, despite reducing peak elevations in intracellular free Ca2+. This injury potentiation may, at least in part, be related to a slowed recovery of intracellular Ca2+ homeostasis, observed after AMPA-kainate receptor activation, but not after NMDA receptor activation or exposure to high K+. The ability of acidic pH to selectively augment AMPA-kainate receptor-mediated excitotoxicity may contribute to the prominent role that these receptors play in selective neuronal death after transient global ischemia.


Subject(s)
Acids/metabolism , Cerebral Cortex/physiology , Extracellular Space/metabolism , Neurons/physiology , Receptors, AMPA/physiology , Anaerobiosis/physiology , Animals , Calcium/metabolism , Cell Death/physiology , Cerebral Cortex/cytology , Excitatory Amino Acid Agonists/pharmacology , Glucose/deficiency , Hydrogen-Ion Concentration , Intracellular Membranes/metabolism , Kainic Acid/pharmacology , Mice/embryology , Neurons/drug effects , Neurons/pathology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
19.
J Biol Chem ; 273(26): 16521-6, 1998 Jun 26.
Article in English | MEDLINE | ID: mdl-9632721

ABSTRACT

Cytokines, including tumor necrosis factor-alpha (TNF-alpha), may elicit cytotoxic response through the sphingomyelin-ceramide signal transduction pathway by activation of sphingomyelinases and the subsequent release of ceramide: the universal lipid second messenger. Treatment of bovine cerebral endothelial cells (BCECs) with TNF-alpha for 16 h followed by cycloheximide (CHX) for 6 h resulted in an increase in ceramide accumulation, DNA fragmentation, and cell death. Application of a cell permeable ceramide analogue C2 ceramide, but not the biologically inactive C2 dihydroceramide, also induced DNA laddering and BCEC death in a concentration- and time-dependent manner. TNF-alpha/CHX-mediated ceramide production apparently is not a result of sphingomyelin hydrolysis because sphingomyelin content does not decrease in this death paradigm. In addition, an acidic sphingomyelinase inhibitor, desipramine, had no effect on TNF-alpha/CHX-induced cell death. However, addition of fumonisin B1, a selective ceramide synthase inhibitor, attenuated TNF-alpha/CHX-induced intracellular ceramide elevation and BCEC death. Together, these findings suggest that ceramide plays at least a partial role in this paradigm of BCEC death. Our results show, for the first time, that ceramide derived from de novo synthesis is an alternative mechanism to sphingomyelin hydrolysis in the BCEC death process initiated by TNF-alpha/CHX.


Subject(s)
Ceramides/biosynthesis , Cerebrovascular Circulation , Cycloheximide/pharmacology , Endothelium, Vascular/drug effects , Protein Synthesis Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cattle , Cell Death/drug effects , Cells, Cultured , Oxidoreductases/antagonists & inhibitors
20.
Science ; 278(5335): 114-7, 1997 Oct 03.
Article in English | MEDLINE | ID: mdl-9311914

ABSTRACT

Apoptosis of mouse neocortical neurons induced by serum deprivation or by staurosporine was associated with an early enhancement of delayed rectifier (IK) current and loss of total intracellular K+. This IK augmentation was not seen in neurons undergoing excitotoxic necrosis or in older neurons resistant to staurosporine-induced apoptosis. Attenuating outward K+ current with tetraethylammonium or elevated extracellular K+, but not blockers of Ca2+, Cl-, or other K+ channels, reduced apoptosis, even if associated increases in intracellular Ca2+ concentration were prevented. Furthermore, exposure to the K+ ionophore valinomycin or the K+-channel opener cromakalim induced apoptosis. Enhanced K+ efflux may mediate certain forms of neuronal apoptosis.


Subject(s)
Apoptosis , Neurons/cytology , Potassium Channels/metabolism , Potassium/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Benzopyrans/pharmacology , Calcium/metabolism , Cerebral Cortex/cytology , Cromakalim , Cycloheximide/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Gadolinium/pharmacology , Mice , N-Methylaspartate/pharmacology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Nifedipine/pharmacology , Patch-Clamp Techniques , Potassium Channels/drug effects , Pyrroles/pharmacology , Staurosporine/pharmacology , Tetraethylammonium , Tetraethylammonium Compounds/pharmacology , Veratridine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...