Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Neurochem Int ; 52(4-5): 887-96, 2008.
Article in English | MEDLINE | ID: mdl-18023503

ABSTRACT

"Ca(2+) paradox" is the phenomenon whereby the intracellular concentration of Ca(2+) paradoxically increases during reperfusion with normal Ca(2+)-containing media after brief exposure to a Ca(2+)-free solution. The present study aims to characterize the Ca(2+) paradox induced cell injury in neuron/astrocyte co-cultures. Prior exposure of the co-cultures to a low Ca(2+) solution for 60 min significantly injured only neurons after reperfusion with a normal Ca(2+) medium for 24h, but astrocytes remained intact. An analysis of the Ca(2+) paradox-induced changes in the intracellular concentration of Na(+) revealed that the concentration in astrocytes increased significantly during the reperfusion episode, resulting in a reversal of the operation of the astrocytic Na(+)-dependent glutamate transporter GLT-1. These results suggested that Ca(2+) paradox-induced accumulation of Na(+) in astrocytes was crucially involved in the excitotoxic neuronal injury resulting from the reversed astrocytic GLT-1 during the reperfusion episode. Previous studies have suggested that Ca(2+) paradox-induced injury in the brain occurs first in astroglial cells and only later in neurons resulting from the prior damage of astrocytes. Here we show that if "Ca(2+) paradox" occurs in the brain, neurons would be the primary target of Ca(2+) paradox-induced cell injury in the central nervous system.


Subject(s)
Astrocytes/drug effects , Calcium/metabolism , Neurons/drug effects , Animals , Astrocytes/metabolism , Blotting, Western , Cell Death/drug effects , Cell Survival/drug effects , Coculture Techniques , Excitatory Amino Acid Transporter 2/biosynthesis , Excitatory Amino Acid Transporter 2/genetics , Extracellular Space/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/metabolism , Immunohistochemistry , Mitogen-Activated Protein Kinase 1/metabolism , Neurons/metabolism , Pregnancy , Rats , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Sodium/metabolism
2.
Neurochem Res ; 32(6): 988-1001, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17401678

ABSTRACT

Spontaneous Ca(2+) oscillations are believed to contribute to the regulation of gene expression. Here we investigated whether and how the dynamics of Ca(2+) oscillations changed after sublethal preconditioning (PC) for PC-induced ischemic tolerance in neuron/astrocyte co-cultures. The frequency of spontaneous Ca(2+) oscillations significantly decreased between 4 and 8 h after the end of PC in both neurons and astrocytes. Treatment with 2-APB, an inhibitor of IP3 receptors, decreased the oscillatory frequency, induced ischemic tolerance and a down-regulation of glutamate transporter GLT-1 contributing to the increase in the extracellular glutamate during ischemia. The expression of GLT-1 is known to be up-regulated by PACAP. Treatment with PACAP38 increased the oscillatory frequency, and antagonized both the PC-induced down-regulation of GLT-1 and ischemic tolerance. These results suggested that the PC suppressed the spontaneous Ca(2+) oscillations regulating the gene expressions of various proteins, especially of astrocytic GLT-1, for the development of the PC-induced ischemic tolerance.


Subject(s)
Brain Ischemia/physiopathology , Calcium Signaling/physiology , Calcium/metabolism , Ischemic Preconditioning , Animals , Astrocytes/physiology , Blotting, Western , Cell Survival , Coculture Techniques , Colforsin/pharmacology , Down-Regulation/physiology , Excitatory Amino Acid Transporter 2/biosynthesis , Excitatory Amino Acid Transporter 2/physiology , Glucose/deficiency , Glutamic Acid/toxicity , Neurons/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Rats
3.
Neurochem Res ; 31(7): 933-43, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16830212

ABSTRACT

During ischemia, the operation of astrocytic/neuronal glutamate transporters is reversed and glutamate and Na(+) are co-transported to the extracellular space. This study aims to investigate whether this reversed operation of glutamate transporters has any functional meanings for astrocytes themselves. Oxygen/glucose deprivation (OGD) of neuron/astrocyte co-cultures resulted in the massive death of neurons, and the cell death was significantly reduced by treatment with either AP5 or DHK. In cultured astrocytes with little GLT-1 expression, OGD produced Na(+) overload, resulting in the reversal of astrocytic Na(+)/Ca(2+)-exchanger (NCX). The reversed NCX then caused Ca(2+) overload leading to the damage of astrocytes. In contrast, the OGD-induced Na(+) overload and astrocytic damage were significantly attenuated in PACAP-treated astrocytes with increased GLT-1 expression, and the attenuation was antagonized by treatment with DHK. These results suggested that the OGD-induced reversal of GLT-1 contributed to the survival of astrocytes themselves by releasing Na(+) with glutamate via reversed GLT-1.


Subject(s)
Astrocytes/metabolism , Brain Ischemia/metabolism , Cell Survival , Excitatory Amino Acid Transporter 2/metabolism , Animals , Coculture Techniques , Immunohistochemistry , Neurons/metabolism , Rats
4.
Neurochem Res ; 31(1): 49-56, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16474996

ABSTRACT

In the brain, prior sublethal ischemia (preconditioning, PC) produces tolerance of neurons to subsequent lethal ischemia. This study aims at elucidating whether and how nitric oxide (NO) produced during PC is involved in the PC-induced ischemic tolerance of neurons in neuron/astrocyte co-cultures. The rise in the extracellular concentration of glutamate during ischemia caused by the reversed uptake of glutamate (Glu) by the astrocytic Glu transporter GLT-1 was markedly suppressed by the prior PC treatment, but the suppression was reversed by treatment with an inhibitor of nitric oxide synthase (NOS) during PC. Immunocytochemical and Western blot analyses demonstrated that the expression of GLT-1 was down-regulated after the PC insult, and this down-regulation was also antagonized by treatment with NOS inhibitors during PC. Here we show that nNOS-derived NO produced during PC was crucial for the down-regulation of astrocytic GLT-1, and this down-regulation coincided with an increased survival rate of neurons.


Subject(s)
Astrocytes/metabolism , Brain Ischemia/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Ischemic Preconditioning , Neurons/metabolism , Nitric Oxide/metabolism , Animals , Astrocytes/cytology , Cell Survival , Cells, Cultured , Coculture Techniques , Down-Regulation , Enzyme Activation , Isoenzymes/metabolism , Neurons/cytology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Rats
5.
Neurochem Res ; 30(9): 1109-16, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16292503

ABSTRACT

In the brain, prior sublethal ischemia (preconditioning, PC) is known to produce tolerance of neurons to subsequent lethal ischemia. This study aims at elucidating what alterations were induced in neurons and/or astrocytes by PC treatment. The rise in the extracellular concentration of glutamate during ischemia was markedly suppressed by the prior PC treatment. Immunocytochemical and Western blot analyses demonstrated that the expression of the astrocytic glutamate transporter GLT-1 was transiently down-regulated after the PC insult. The PC insult possibly suppressed the neuron-derived factors up-regulating GLT-1. Here we show that PC-induced down-regulation of GLT-1 is crucial for the increased neuronal resistance to subsequent severe ischemic insult.


Subject(s)
Astrocytes/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Ischemic Preconditioning , Neurons/metabolism , Animals , Astrocytes/cytology , Brain Ischemia/metabolism , Cell Survival , Cells, Cultured , Coculture Techniques , Down-Regulation , Embryo, Mammalian/anatomy & histology , Glutamic Acid/metabolism , Neurons/cytology , Rats
6.
Glia ; 49(3): 349-59, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15538756

ABSTRACT

Sublethal ischemia leads to increased tolerance against subsequent prolonged cerebral ischemia in vivo. In the present study, we investigated the roles of the astrocytic glutamate (Glu) transporter GLT-1 in preconditioning (PC)-induced neuronal ischemic tolerance in cortical neuron/astrocyte co-cultures. Ischemia in vitro was simulated by subjecting cultures to both oxygen and glucose deprivation (OGD). A sublethal OGD (PC) increased the survival rate of neurons significantly when cultures were exposed to a lethal OGD 24 h later. The extracellular concentration of Glu increased significantly during PC, and treatment with an inhibitor of N-methyl-D-actetate (NMDA) receptors significantly reversed the PC-induced ischemic tolerance of neurons, suggesting that the increase in extracellular concentration of Glu during PC was critical to the development of PC-induced neuronal ischemic tolerance via the activation of NMDA receptors. Treatment with a GLT-1 blocker during PC suppressed this increase in Glu significantly, and antagonized the PC-induced neuronal ischemic tolerance. This study suggested that the reversed operation of GLT-1 was crucial to the development of neuronal ischemic tolerance.


Subject(s)
Astrocytes/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Neurons/metabolism , Animals , Astrocytes/cytology , Cell Hypoxia/physiology , Cell Survival/physiology , Coculture Techniques , Glucose/deficiency , Glutamic Acid/metabolism , Neurons/cytology , Rats
7.
Neurochem Res ; 29(4): 797-804, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15098944

ABSTRACT

The present study investigated the roles of nitric oxide (NO) in preconditioning (PC)-induced neuronal ischemic tolerance in cortical cultures. Ischemia in vitro was simulated by subjecting cultures to both oxygen and glucose deprivation (OGD). A sublethal OGD (PC) significantly increased the survival rate of neurons when cultures were exposed to a lethal OGD 24 h later. Both the inhibition of nitric oxide synthase (NOS) and scavenging of NO during PC significantly attenuated the PC-induced neuronal tolerance. In addition, exposure to an NO donor emulated the PC. In contrast, the inhibition of NOS and the scavenging of NO during lethal OGD tended to increase the survival rate of neurons. This study suggested that NO produced during ischemia was fundamentally toxic, but critical to the development of PC-induced neuronal tolerance.


Subject(s)
Ischemic Preconditioning , Nitric Oxide/physiology , Animals , Cells, Cultured , Immunohistochemistry , Nitric Oxide/toxicity , Rats
SELECTION OF CITATIONS
SEARCH DETAIL