Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta ; 1647(1-2): 220-4, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12686136

ABSTRACT

Lysyl oxidase (LOX) and four lysyl oxidase-like proteins, LOXL, LOXL2, LOXL3 and LOXL4, each contain a copper binding site, conserved lysyl and tyrosyl residues that may contribute to quinone co-factor formation, and a cytokine receptor-like domain. Each protein differs mainly in their N-terminal sequence, which may confer individual functions. Processing of the LOX proteins by BMP-1 and possibly other mechanisms may result in multiple functional forms. Splicing, reported for LOXL3, may also generate additional variants with unique functions. Each LOX, with its individual, developmentally regulated tissue and cell-specific expression and localization, results in a complex structural and functional variation for the LOX amine oxidases. The presence of only two LOX-like proteins in Drosophila, each with distinct spatial and temporal expression, allows for the assignment of individual function to one of these amine oxidases. Comparative expression analysis of each LOX protein is presented to help determine their functional significance.


Subject(s)
Amino Acid Oxidoreductases/chemistry , Protein-Lysine 6-Oxidase/chemistry , Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/physiology , Animals , Drosophila/enzymology , Gene Expression Regulation, Developmental , Mice , Myocardium/enzymology , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/physiology
2.
Cell Mol Biol (Noisy-le-grand) ; 49(8): 1241-7, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14983993

ABSTRACT

Transient global cerebral ischemia leads to delayed neuronal cell death in the hippocampal CA1, caudate putamen and neocortex. If preischemic hyperglycemia exists, the same duration of ischemia recruits additional brain structures, such as dentate gyrus to become damaged. The objective of the present study is to determine whether activation of mitogen-activated protein kinases (MAPKs) plays a role in hyperglycemia-mediated ischemic neuronal damage. Using phopho-specific antibodies against c-jun NH2-terminal kinase (JNK) and p38 MAPK, we studied activation of these two MAPKs in ischemia-vulnerable neocortex and ischemia-resistant dentate gyrus in rats subjected to 15 min of forebrain ischemia and followed by 0.5, 1 and 3 hr of recirculation under normo- and hyperglycemic conditions. The results showed that levels of phosphorylated JNK increased in both normo- and hyperglycemic brains following blood reperfusion for 0.5 hr and persisted up to 3 hr in the neocortex but not in the dentate gyrus, implying JNK may play a role in mediating neuronal cell death after ischemia. However, since hyperglycemia did not further increase phospho-JNK, JNK may not contribute to the detrimental effect of hyperglycemia on neuronal cell death. The amount of phospho-p38 was not altered by ischemia under both normo- and hyperglycemic conditions, suggesting that p38 MAPK may not play a major role in mediating neuronal damage in these two structures.


Subject(s)
Brain Ischemia/pathology , Hyperglycemia/pathology , Mitogen-Activated Protein Kinases/metabolism , Neurons/pathology , Animals , Cell Death , Dentate Gyrus/enzymology , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Enzyme Activation , JNK Mitogen-Activated Protein Kinases , Male , Neocortex/enzymology , Neocortex/metabolism , Neocortex/pathology , Neurons/enzymology , Neurons/metabolism , Phosphorylation , Rats , Rats, Wistar , p38 Mitogen-Activated Protein Kinases
3.
Cell Mol Biol (Noisy-le-grand) ; 49(8): 1249-52, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14983994

ABSTRACT

Recent studies have demonstrated that disodium 2,4-disulfophenyl-N-tert-butylnitrone (NXY-059), a novel nitrone with free radical trapping properties, has a considerable neuroprotective effect against cerebral ischemic injury. The mechanisms of its action have not been fully defined. In order to evaluate whether NXY-059 exerts its protective effects by inhibiting the release of cytochrome c, a key initiator of programmed cell death pathway, we have studied the effects of NXY-059 on reducing infarct volume and on inhibiting cytochrome c release from the mitochondria after transient focal cerebral ischemia. Wistar rats were subjected to 2 hr of middle cerebral artery occlusion and perfusion-fixed after 4, 6, 12, and 24 hr of reperfusion. NXY-059 (30 mg/kg) was i.v. injected 1 hr after reperfusion and followed immediately by 30 mg/kg/hr continuous i.v. infusion for the entire reperfusion period. The results showed that NXY-059 reduced infarct volume from 37.2% to 12.5% (p<0.0001). Immunocytochemistry demonstrated that the release of cytochrome c increased at 6 hr, peaked at 12 and 24 hr of reperfusion. NXY-059 treatment prevented ischemia-induced cytochrome c release. NXY-059 may reduce ischemic brain damage through suppressing the cell death pathway that is initiated by cytochrome c release.


Subject(s)
Cytochromes c/drug effects , Ischemic Attack, Transient/drug therapy , Nitrogen Oxides/pharmacology , Animals , Benzenesulfonates , Cell Death , Cerebral Infarction/prevention & control , Cytochromes c/metabolism , Free Radical Scavengers/pharmacology , Free Radical Scavengers/therapeutic use , Male , Mitochondria/drug effects , Mitochondria/pathology , Nitrogen Oxides/therapeutic use , Rats , Rats, Wistar , Reperfusion
4.
Free Radic Biol Med ; 31(10): 1191-7, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11705697

ABSTRACT

It has been documented that alpha-phenyl-N-tert-butyl-nitron (PBN) possesses a potent neuroprotective effect when administered after transient focal cerebral ischemia. However, contradicting results were reported regarding its effect in transient global ischemia. To further elucidate the mechanism of PBN action, we have studied the effect of PBN on animal survival, histopathological outcome, and activation of caspase-3 following 30 min of global ischemia in vehicle- and PBN-treated rats. The results showed that 30 min of global ischemia was such a severe insult that no animal could survive beyond 2 d of reperfusion. Histopathological evaluation showed severe tissue edema and microinfarct foci in the neocortex and thalamus. Close to 100% damage was observed in the stratum and hippocampal CA1, CA3, and dentate gyrus subregions. Postischemic PBN treatment significantly enhanced animal survival and reduced damage in the neocortex, thalamus, and hippocampus. Immunohistochemistry demonstrated that caspase-3 was activated following ischemia in the striatum and the neocortex. PBN suppressed the activation of caspase-3 in both structures. It is concluded that PBN is a potent neuroprotectant against both focal and global ischemia; besides its function as a free radical scavenger, PBN may reduce ischemic brain damage by blocking cell death pathways that involve caspase-3 activation.


Subject(s)
Brain Ischemia/drug therapy , Caspase Inhibitors , Neuroprotective Agents/therapeutic use , Nitrogen Oxides/therapeutic use , Animals , Brain Ischemia/pathology , Caspase 3 , Cyclic N-Oxides , Disease Models, Animal , Male , Rats , Rats, Wistar , Spin Trapping , Survival Rate
5.
Brain Res ; 912(2): 203-5, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11532437

ABSTRACT

A recent study reported that hyperglycemia of a brief duration worsens, and of long duration reduces, ischemic brain damage. To test whether this is a valid conception, we induced 10 min of transient forebrain ischemia, recorded postischemic seizures, and evaluated brain morphology. The results showed that administration of glucose 2 h before ischemia aggravated brain damage, induced seizures, and caused animal death in the same manner as was previously observed when glucose was given 30 min before ischemia. Thus, the conclusion that the influence of glucose on an ischemic transient is dependent upon the duration of hyperglycemia is unsubstantiated.


Subject(s)
Brain Injuries/drug therapy , Brain Ischemia/drug therapy , Brain/drug effects , Cerebral Infarction/drug therapy , Glucose/pharmacology , Reperfusion Injury/drug therapy , Animals , Brain/pathology , Brain/physiopathology , Brain Injuries/pathology , Brain Injuries/physiopathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebral Infarction/pathology , Cerebral Infarction/physiopathology , Drug Administration Schedule , Male , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Rats , Rats, Wistar , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Survival Rate
6.
J Cereb Blood Flow Metab ; 21(5): 568-76, 2001 May.
Article in English | MEDLINE | ID: mdl-11333367

ABSTRACT

Previous histopathologic results have suggested that one mechanism whereby hyperglycemia (HG) leads to exaggerated ischemic damage involves fragmentation of DNA. DNA fragmentation in normoglycemia (NG) and HG rats subjected to 30 minutes of forebrain ischemia was studied by terminal deoxynucleotidyl transferase mediated DNA nick-labeling (TUNEL) staining, by pulse-field gel electrophoresis (PFGE), and by ligation-mediated polymerase chain reaction (LM-PCR). High molecular weight DNA fragments were detected by PFGE, whereas low molecular weight DNA fragments were detected using LM-PCR techniques. The LM-PCR procedure was performed on DNA from test samples with blunt (without Klenow polymerase) and 3'-recessed ends (with Klenow polymerase). In addition, cytochrome c release and caspase-3 activation were studied by immunocytochemistry. Results show that HG causes cytochrome c release, activates caspase-3, and exacerbates DNA fragments induced by ischemia. Thus, in HG rats, but not in control or NGs, TUNEL-stained cells were found in the cingulate cortex, neocortex, thalamus, and dorsolateral crest of the striatum, where neuronal death was observed by conventional histopathology, and where both cytosolic cytochrome c and active caspase-3 were detected by confocal microscopy. In the neocortex, both blunt-ended and stagger-ended fragments were detected in HG, but not in NG rats. Electron microscopy (EM) analysis was performed in the cingulate cortex, where numerous TUNEL-positive neurons were observed. Although DNA fragmentation was detected by TUNEL staining and electrophoresis techniques, EM analysis failed to indicate apoptotic cell death. It is concluded that HG triggers a cell death pathway and exacerbates DNA fragmentation induced by ischemia.


Subject(s)
DNA Fragmentation , Hyperglycemia/pathology , Ischemic Attack, Transient/pathology , Animals , Apoptosis , Caspase 3 , Caspases/metabolism , Corpus Striatum/pathology , Cytochrome c Group/metabolism , Dentate Gyrus/pathology , Enzyme Activation , Hippocampus/pathology , Hyperglycemia/physiopathology , In Situ Nick-End Labeling , Ischemic Attack, Transient/physiopathology , Male , Microscopy, Electron , Neocortex/pathology , Neurons/pathology , Rats , Rats, Wistar , Thalamus/pathology
7.
Brain Res ; 899(1-2): 148-58, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11311876

ABSTRACT

The two immunosuppressants, cyclosporin A (CsA) and FK506, when given 1 and 3 h after the start of reperfusion following 2 h of middle cerebral artery (MCA) occlusion, reduce infarct volume to 30% of control. This suggests a common effect, e.g. one due to suppression of the activation of calcineurin. However, when given by the intracarotid (i.c.) route after only 5 min of recirculation CsA, but not FK506, reduced infarct volume even further, to 10% of control. This was attributed to the fact that CsA, but not FK506, block the in vitro assembly of a mitochondrial permeability transition (MPT) pore. The present experiments were undertaken to further characterize the anti-ischemic effect of CsA, when given i.c. 5 min after recirculation and to explore why CsA, when given at that time, is more efficacious than FK506. It was established that the i.c. administration of CsA in a dose of 10 mg/kg increased the tissue concentration of CsA 2- to 3-fold, when compared to the i.v. administration. CsA proved to be effective in reducing infarct volume even when the tissue damage was assessed by histopathology after 7 days of recovery. MCA occlusion of 2 h duration caused a sustained decrease in the phosphorylation Akt at threonine 308. Since this down regulation of Akt was prevented by CsA, the results suggested a link between dephosphorylaltion of Bad, and cell death. Interestingly FK506 did not prevent down regulation of Akt, it thus seems unlikely that the anti-ischemic effect of CsA is related to its association with cytosolic cyclophilin.


Subject(s)
Cyclosporine/pharmacology , Down-Regulation/drug effects , Immunosuppressive Agents/pharmacology , Ischemic Attack, Transient/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Tacrolimus/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Down-Regulation/physiology , Male , Phosphorylation , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar
8.
Brain Res ; 896(1-2): 69-76, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11277975

ABSTRACT

The mechanisms underlying the aggravating effect of hyperglycemia on brain damage are still elusive. The present study was designed to test our hypothesis that hyperglycemia-mediated damage is caused by mitochondrial dysfunction with mitochondrial release of cytochrome c (cyt c) to the cytoplasm, which leads to activation of caspase-3, the executioner of cell death. We induced 15 min of forebrain ischemia, followed by 0.5, 1, and 3 h of recirculation in sham, normoglycemic and hyperglycemic rats. Release of cyt c was observed in the neocortex and CA3 in hyperglycemic rats after only 0.5 h of reperfusion, when no obvious neuronal damage was observed. The release of cyt c persisted after 1 and 3 h of reperfusion. Activation of caspase-3 was observed after 1 and 3 h of recovery in hyperglycemic animals. No cyt c release or caspase-3 activation was observed in sham-operated controls while a mild increase of cyt c was observed in normoglycemic ischemic animals after 1 and 3 h of reperfusion. The findings that there is caspase activation and cyt c relocation support a notion that the biochemical changes that constitute programmed cell death occur after ischemia and contribute, at least in part, to hyperglycemia-aggravated ischemic neuronal death.


Subject(s)
Caspases/metabolism , Cytochrome c Group/metabolism , Hyperglycemia/metabolism , Ischemic Attack, Transient/metabolism , Animals , Blotting, Western , Caspase 3 , Cell Death/physiology , Cytochrome c Group/analysis , In Situ Nick-End Labeling , Male , Mitochondria/enzymology , Neurons/cytology , Neurons/enzymology , Prosencephalon/blood supply , Prosencephalon/cytology , Prosencephalon/metabolism , Rats , Rats, Wistar
9.
Brain Res ; 890(2): 306-13, 2001 Feb 02.
Article in English | MEDLINE | ID: mdl-11164797

ABSTRACT

A recent study showed that a single intracarotid arterial injection of cyclosporin A (CsA) can dramatically reduce infarct volume in rats subjected to transient focal ischemia. The present experiments were undertaken to investigate whether intracarotid arterial injection of CsA reduces brain damage after global ischemia. Since hypothermia is also an efficacious factor in preventing ischemic brain damage, in the second part of the experiments we tested whether a combination of hypothermia and CsA would provide additional brain protection. Global ischemia of a 30-min duration was induced in the rat. CsA (10 mg/kg) was injected into the carotid artery immediately after reperfusion. Hypothermia was instituted after ischemia by allowing spontaneous head temperature to fall to 30-32 degrees C, while body temperature was upheld at 37 degrees C. The results demonstrated that vehicle-treated animals could not survive beyond 1-2 days after reperfusion, and the histopathological outcome in a separate group of rats perfusion-fixed after 1 day reperfusion showed 80-100% brain damage in the caudoputamen, and in the hippocampal CA1, CA3, CA4 and dentate gyrus subregions. Microinfarction and grade 3 damage were frequently observed in the cingulate and parietal cortex and in the thalamus. CsA moderately prolonged animal survival to 3 days after reperfusion and reduced brain damage to grade 2 in the cortical areas and the thalamus. Hypothermia further increased animal survival to at least 6 days after reperfusion and reduced brain damage to 30% in the caudoputamen, to close to zero in the CA3, CA4, and dentate gyrus, and to grade 1-2 in the cortical areas and the thalamus. The combination of hypothermia and CsA did not give additional protection.


Subject(s)
Brain Infarction/drug therapy , Brain Ischemia/drug therapy , Cyclosporine/pharmacology , Hypothermia, Induced , Immunosuppressive Agents/pharmacology , Neuroprotective Agents/pharmacology , Animals , Blood Glucose/physiology , Brain Infarction/pathology , Brain Infarction/physiopathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cardiovascular Physiological Phenomena , Carotid Arteries/surgery , Injections, Intra-Arterial , Male , Rats , Rats, Wistar , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Respiratory Physiological Phenomena , Survival Rate , Time Factors
10.
Neurobiol Dis ; 8(1): 127-35, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11162246

ABSTRACT

The present study was undertaken to investigate whether extracellular signal-regulated kinase (ERK) was involved in mediating hyperglycemia-exaggerated cerebral ischemic damage. Phosphorylation of ERK 1/2 was studied by immunocytochemistry and by Western blot analyses. Rats were subjected to 15 min of forebrain ischemia, followed by 0.5, 1, and 3 h of reperfusion under normoglycemic and hyperglycemic conditions. The results showed that in normoglycemic animals, moderate phosphorylation of ERK 1/2 was transiently induced after 0.5 h of recovery in cingulate cortex and in dentate gyrus, returning to control values thereafter. In hyperglycemic animals, phosphorylation of ERK 1/2 was markedly increased in the cingulate cortex and dentate gyrus after 0.5 h of recovery, the increases being sustained for at least 3 h after reperfusion. Hyperglycemia also induced phosphorylation of ERK 1/2 in the hippocampal CA3 sector but not in the CA1 area. Thus, the distribution of phospho-ERK 1/2 coincides with hyperglycemia-recruited damage structures. The results suggest that hyperglycemia may influence the outcome of an ischemic insult by modulating signal transduction pathways involving ERK 1/2.


Subject(s)
Hyperglycemia/metabolism , Ischemic Attack, Transient/metabolism , Mitogen-Activated Protein Kinases/metabolism , Animals , Blood Glucose/metabolism , Blotting, Western , Brain/pathology , Cell Nucleus/metabolism , Hyperglycemia/enzymology , Hyperglycemia/pathology , Immunohistochemistry , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/pathology , Male , Microscopy, Confocal , Phosphorylation , Rats , Rats, Wistar , Subcellular Fractions/metabolism , Up-Regulation/drug effects
11.
J Cereb Blood Flow Metab ; 20(11): 1550-6, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11083229

ABSTRACT

In the current study, the temporal and regional changes of the transcription factor cyclic adenosine monophosphate response element binding protein (CREB) were investigated in rat brains subjected to 30 minutes of hypoglycemic coma followed by varied periods of recovery using Western blot and confocal microscopy. The total amount of CREB was not altered in any area examined after coma. The level of the phosphorylated form of CREB decreased during coma but rebounded after recovery. In the relatively resistant areas, such as the inner layers of the neocortex and the inner and outer blades of the dentate gyms (DG), phospho-CREB increased greater than the control level after 30 minutes of recovery and continued to increase up to 3 hours of recovery. In contrast, little or no increase of phospho-CREB was observed during the recovery period in the outer layers of the neocortex and at the tip of the DG, that is, regions that are selectively vulnerable to hypoglycemic insults. The current findings suggest that a neuroprotective signaling pathway may be more activated in the resistant regions than in the vulnerable ones after hypoglycemic coma.


Subject(s)
Brain/metabolism , Coma/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP/metabolism , Hypoglycemia/metabolism , Animals , Antibody Specificity , Blotting, Western , Coma/etiology , Cyclic AMP Response Element-Binding Protein/analysis , Cyclic AMP Response Element-Binding Protein/immunology , Fluorescent Antibody Technique , Hippocampus/chemistry , Hippocampus/metabolism , Hypoglycemia/complications , Male , Microscopy, Confocal , Neocortex/chemistry , Neocortex/metabolism , Neostriatum/chemistry , Neostriatum/metabolism , Phosphorylation , Rats , Rats, Wistar , Signal Transduction/physiology
12.
Brain Res ; 876(1-2): 191-5, 2000 Sep 08.
Article in English | MEDLINE | ID: mdl-10973608

ABSTRACT

We analyzed both total Akt-1 and phosphorylation of Akt-1 at residues Ser473 and Thr308 (phospho-Akt-1(Ser474) and phospho-Akt-1(Thr308), respectively) in the outer and inner layers of cortex following 30 min of hypoglycemic coma by Western blot analyses and confocal microscopy. The total amount of Akt-1 was not altered in any area examined. Phospho-Akt-1(Ser474), however, increased significantly in both layers of cortex at 0 and 30 min of recovery, but returned to control level at 3 h of recovery. In the vulnerable area (outer layer of cortex), no upregulation of phospho-Akt-1(Thr308) was observed at any time points examined. In the resistant area like inner layer of cortex, however, phospho-Akt-1(Thr308) was significantly over the control level at 3 h of recovery. Confocal microscopy result indicates that most of phospho-Akt-1(Thr308) had already moved into nucleus at 3 h of recovery. Our results suggest that Akt-1, when phosphorylated at Thr308, may play a protective role for neurons in the resistant regions of the brain.


Subject(s)
Brain/metabolism , Coma/etiology , Coma/metabolism , Hypoglycemia/complications , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , Amino Acid Sequence/genetics , Animals , Blotting, Western , Male , Microscopy, Confocal , Phosphorylation , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar , Time Factors , Tissue Distribution
13.
Exp Neurol ; 165(1): 153-63, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10964494

ABSTRACT

Cyclosporin A (CsA) has been shown to be efficacious in protecting against ischemic injury after short periods (5 to 10 min) of forebrain ischemia. The present experiments were undertaken to study if a long period of forebrain ischemia (30 min), induced at a brain temperature of 37 degrees C, is compatible with survival and if the brain damage incurred can be ameliorated by CsA. The results showed that animals subjected to 30 min of forebrain ischemia at a brain temperature of 37 degrees C failed to survive after the first 24 h of recovery and showed extensive neuronal necrosis in all selectively vulnerable regions after 1 day of survival. CsA, when injected in combination with an intracerebral lesion to open the blood-brain barrier, markedly prolonged the survival time. CsA-injected animals also showed amelioration of histological lesions, an effect that was sustained for at least 4 days. Experiments with mitochondria isolated from the neocortex and hippocampus showed that state 3 respiratory rates decreased during ischemia, recovered after 1 and 3 h of recirculation, and then showed a secondary decline at 6 h. Administration of CsA prevented this secondary decline. Measurements of neocortical cerebral blood flow showed that there was no secondary hypoperfusion prior to secondary mitochondrial dysfunction, implying that changes in blood flow may not be responsible for the rapidly developing, secondary brain damage. The results thus demonstrate that if brain temperature is upheld at 37 degrees C, a 30-min period of ischemia is not compatible with survival after the first day of recovery, and gross histopathological damage develops within that period. CsA was efficacious in prolonging animal survival, ameliorating brain damage, and preventing the secondary mitochondrial dysfunction. Since CsA blocks the mitochondrial permeability transition pore its action may, at least in part, be on mitochondrial integrity and function.


Subject(s)
Brain Damage, Chronic/mortality , Brain Damage, Chronic/pathology , Cyclosporine/pharmacology , Ischemic Attack, Transient/complications , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Animals , Cerebrovascular Circulation/drug effects , Ischemic Attack, Transient/physiopathology , Male , Mitochondria/metabolism , Oxygen Consumption/drug effects , Rats , Rats, Wistar , Time Factors
14.
Neurochem Res ; 25(5): 661-7, 2000 May.
Article in English | MEDLINE | ID: mdl-10905628

ABSTRACT

In this study, we explored if a 30 minute period of hypoglycemic coma yields damage which shows some features associated with apoptosis. To that end, we induced insulin-hypoglycemic coma of 30 min duration, and studied brain tissues after the coma period, and after recovery period of 30 min, 3 h, and 6 h. Histopathological data confirmed neuronal damage in all of the vulnerable neuronal populations. Release of cytochrome c (cyt c), assessed by Western Blot, was observed in the neocortex and caudoputamen after 3 and 6 h of recovery. In these regions, the caspase-like activity increased above control after 6 h of recovery. By laser-scanning confocal microscopy, a clear expression of Bax was observed after 30 min of coma in the superficial layers of the neocortex, reaching a peak after 30 min of recovery. Punctuate immunolabeling surrounding nuclei in soma and dendrites in cortical pyramidal neurons likely represents mitochondria, which suggests that Bax protein assembled at the surface of mitochondria in vulnerable neocortical neurons. It is concluded that although previous morphological data have suggested that cells die by necrosis, neuronal damage after hypoglycemic coma shows some features of apoptosis.


Subject(s)
Apoptosis , Brain/pathology , Hypoglycemia/pathology , Insulin Coma/pathology , Neurons/pathology , Animals , Caspase 3 , Caspases/analysis , Cytochrome c Group/analysis , Electroencephalography , Hypoglycemia/physiopathology , Insulin Coma/physiopathology , Male , Necrosis , Neurons/physiology , Rats , Rats, Wistar , Time Factors
15.
Stroke ; 31(1): 183-92, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10625736

ABSTRACT

BACKGROUND AND PURPOSE: An increase in serum glucose at the time of acute ischemia has been shown to adversely affect prognosis. The mechanisms for the hyperglycemia-exacerbated damage are not fully understood. The objective of this study was to determine whether hyperglycemia leads to enhanced accumulation of extracellular concentrations of excitatory amino acids and whether such increases correlate with the histopathological outcome. METHODS: Rats fasted overnight were infused with either glucose or saline 45 minutes before the induction of 15 minutes of forebrain ischemia. Extracellular glutamate, glutamine, glycine, taurine, alanine, and serine concentrations were measured before, during, and after ischemia in both the hippocampus and the neocortex in both control and hyperglycemic animals. The histopathological outcome was evaluated by light microscopy. RESULTS: There was a significant increase in extracellular glutamate levels in the hippocampus and cerebral cortex in normoglycemic ischemic animals. The increase in glutamate levels in the cerebral cortex, but not in the hippocampus, was significantly higher in hyperglycemic animals than in controls. Correspondingly, exaggerated neuronal damage was observed in neocortical regions in hyperglycemic animals. CONCLUSIONS: The present results demonstrate that, at least in the neocortex, preischemic hyperglycemia enhances the accumulation of extracellular glutamate during ischemia, providing a tentative explanation for why neuronal damage is exaggerated.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Glutamic Acid/metabolism , Hyperglycemia/metabolism , Hyperglycemia/physiopathology , Prosencephalon/blood supply , Alanine/metabolism , Animals , Glutamine/metabolism , Glycine/metabolism , Male , Rats , Rats, Wistar , Serine/metabolism , Taurine/metabolism
16.
Free Radic Biol Med ; 27(9-10): 1033-40, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10569636

ABSTRACT

Preischemic hyperglycemia is known to aggravate brain damage resulting from transient ischemia. In the present study, we explored whether this aggravation is preceded by an enhanced formation of reactive oxygen species (ROS) during the early reperfusion period. To that end, normo- and hyperglycemic rats were subjected to 15 min of forebrain ischemia and allowed recovery periods of 5, 15, and 60 min. Sodium salicylate was injected intraperitoneally in a dose of 100 mg/kg, and tissues were sampled during recirculation to allow analyses of salicylic acid (SA) and its hydroxylation products, 2,3- and 2,5-dihydroxybenzoate (DHBA). Tissue sampled from thalamus and caudoputamen in normoglycemic animals failed to show an increase in 2,3- or 2,5-DHBA after 5 and 15 min of recirculation. However, such an increase was observed in the neocortex after 60 min of recirculation, with a suggested increase in the hippocampus as well. Hyperglycemia had three effects. First, it increased 2,5-DHBA in the thalamus and caudoputamen to values exceeding normoglycemic ones after 15 min of recirculation. Second, it increased basal values of 2,5- and total DHBA in the neocortex. Third, it increased the 60-min values for 2,5- and total DHBA in the hippocampus. These results hint that, at least in part, hyperglycemia may aggravate damage by enhancing basal- and ischemia-triggered production of ROS.


Subject(s)
Brain Ischemia/complications , Brain Ischemia/metabolism , Brain/metabolism , Gentisates , Hydroxyl Radical/metabolism , Hyperglycemia/complications , Hyperglycemia/metabolism , Animals , Brain Injuries/etiology , Brain Injuries/metabolism , Hydroxybenzoates/metabolism , Male , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Salicylic Acid/metabolism , Tissue Distribution
17.
Exp Neurol ; 158(1): 242-53, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10448438

ABSTRACT

It has previously been shown that hypothermia markedly reduces cellular release of the excitatory amino acid glutamate and ameliorates ischemic damage. Based on extensive data showing that preischemic hyperglycemia exaggerates brain damage due to transient forebrain ischemia we posed the question whether glutamate release during ischemia in hyperglycemic rats is attenuated or prevented by induced hypothermia, and if such attenuation/prevention correlates with amelioration of the characteristic brain damage observed in hyperglycemic subjects. The experiments were performed in rats subjected to a 15-min period of forebrain ischemia, plasma glucose concentration being maintained at approximately 5 mM (control) or approximately 20 mM (hyperglycemia) prior to ischemia. Extracellular amino acid concentrations were measured by HPLC techniques on microdialysis samples which were collected from left dorsal hippocampus and right neocortex, and tissue damage was assessed by histopathology. Hypothermia (30 degrees C), which was induced 45 min prior to ischemia, reduced the neuronal damage not only in the ischemia-vulnerable regions but also in the normally ischemia-resistant areas that are recruited in the damage process in hyperglycemic subjects. The extracellular glutamate concentration was markedly increased in response to the ischemic insult in normothermic-normoglycemic animals. The concentration of glutamate was further increased in normothermic-hyperglycemic animals. Hypothermia inhibited the rise in glutamate concentrations, as well as in the concentrations of other excitatory and inhibitory amino acids. It is discussed whether hypothermia reduces the hyperglycemia-mediated damage by inhibiting extracellular glutamate release during an ischemic transient.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain/pathology , Excitatory Amino Acids/metabolism , Extracellular Space/metabolism , Glutamic Acid/metabolism , Hyperglycemia/metabolism , Hypothermia/metabolism , Prosencephalon/blood supply , Prosencephalon/metabolism , Animals , Brain Ischemia/complications , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Glutamic Acid/analysis , Hippocampus/metabolism , Hippocampus/pathology , Male , Prosencephalon/chemistry , Rats , Rats, Wistar , Seizures/etiology , Time Factors , gamma-Aminobutyric Acid/analysis
18.
Neurosci Lett ; 266(2): 121-4, 1999 May 07.
Article in English | MEDLINE | ID: mdl-10353342

ABSTRACT

Status epilepticus (SE), i.e. ongoing seizures of more than 30 min duration, gives rise to bilateral pan-necrotic lesions of the substantia nigra, pars reticulata (SNPR). These are known to be preceded by an initial increase, followed by a depression of metabolic rate, and by failure of the bioenergetic state, suggesting mitochondrial dysfunction. We have previously shown that the spin trap alpha-phenyl-N-tert-butyl nitrone (PBN) prevents the lesions caused by 45 min of SE from occurring, in spite of ongoing seizure activity. In this article, we demonstrate that PBN, given 30 min before seizure induction, reduces or prevents the decrease in ATP concentration and adenylate energy charge, without significantly reducing the amount of lactate accumulated, or the decrease in intracellular pH (pHi). The results suggest that the spin trap nitrone preserves the structural and functional integrity of SNPR neurons by protecting the mitochondria against oxidative damage.


Subject(s)
Convulsants/toxicity , Energy Metabolism/drug effects , Flurothyl/toxicity , Nitrogen Oxides/pharmacology , Status Epilepticus/drug therapy , Substantia Nigra/drug effects , Animals , Cyclic N-Oxides , Free Radical Scavengers/pharmacology , Male , Rats , Rats, Wistar , Spin Labels , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Substantia Nigra/metabolism
19.
Brain Res ; 812(1-2): 216-26, 1998 Nov 23.
Article in English | MEDLINE | ID: mdl-9813336

ABSTRACT

The immunosuppressant drug cyclosporin A (CsA) is considered to be inherently protective in conditions of ischemia, e.g. in hepatic and cardiac tissue. However, investigations of effects of CsA on neuronal tissue have been contradictory, probably because the blood-brain barrier (BBB) is virtually impermeable to CsA. In the present study, we exploited the finding that the insertion of a syringe needle into brain parenchyma obviously disrupts the BBB and allows influx of CsA, and explored whether CsA, given as intraperitoneal injections daily for 1 week before and 1 week after forebrain ischemia of 7 or 10 min duration, ameliorates the damage incurred to the hippocampal CA 1 sector. In other experiments, the needle insertion and the first i.p. injection of CsA were made 30 min after the start of recirculation, with continued daily administration of CsA during the postinsult week. In animals which were injected with CsA in daily doses of 10 mg kg-1, but in which no needle was inserted, the drug failed to ameliorate CA1 damage, whether the ischemia had a duration of 7 or 10 min. Likewise, needle insertion had no effect on CA1 damage if CsA was not administered. In contrast, when CsA was given to animals with a needle insertion, CA1 damage was dramatically ameliorated, whether treatment was initiated 1 week before ischemia, or 30 min after the start of recirculation. The effect of CsA seemed larger than that of any other drug proposed to have an anti-ischemic effect in forebrain/global ischemia. Injection of tritiated CsA in one animal with BBB disruption lead to detectable radioactivity throughout the ventricular system, suggesting a generalised increase of the entry of CsA across the BBB. The results demonstrate that immunosuppressants of the type represented by CsA markedly ameliorate delayed neuronal damage after transient forebrain ischemia, provided that they can pass the BBB. It is discussed whether the effect of the drug is one involving calcineurin, a protein phosphatase, or if CsA counteracts a permeability transition of the inner mitochondrial membrane, assumed to occur in response to adverse conditions, e.g. gradual accumulation of Ca2+ in the mitochondria in the postischemic period.


Subject(s)
Cyclosporine/therapeutic use , Immunosuppressive Agents/therapeutic use , Ischemic Attack, Transient/drug therapy , Neuroprotective Agents/therapeutic use , Prosencephalon/drug effects , Analysis of Variance , Animals , Blood-Brain Barrier/physiology , Cyclosporine/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Neuroprotective Agents/pharmacokinetics , Prosencephalon/blood supply , Rats , Treatment Outcome
20.
Neurosci Lett ; 253(3): 191-4, 1998 Sep 11.
Article in English | MEDLINE | ID: mdl-9792243

ABSTRACT

Reperfusion after transient focal ischemia of 2 h duration is followed by secondary bioenergetic failure after 4 h of reperfusion. The objective of the present study was to explore whether or not this secondary deterioration is due to secondary microcirculatory compromise. Normal fasted rats were subjected to 2 h of MCA occlusion and allowed reperfusion for 2, 4, 6 and 8 h. At predetermined reperfusion times, rats were injected with Evans blue and decapitated. Capillary patency was determined using a fluorescent double-staining technique. No capillary perfusion deficits were detected in the ischemic neocortical penumbra, neocortical focus or striatal focus. We concluded that the secondary deterioration of bioenergetic state is not due to microcirculatory compromise. Since hyperglycemic animals show pan-necrotic lesions, a hyperglycemic group was added at 8 h of reperfusion to test if the adverse effect of hyperglycemia on ischemic damage is related to capillary compromise. The results showed that, in hyperglycemic rats, capillary perfusion in the striatal focus was compromised after 8 h of recirculation following 2 h of MCA occlusion. It is concluded that when normoglycemic rats are subjected to 2 h of MCA occlusion, capillary patency is not affected during the first 4-6 h of reflow. At 8 h of reflow, though, particularly in hyperglycemic rats, microcirculation is compromised in the caudoputamenal focus, probably reflecting infarction.


Subject(s)
Capillaries/physiology , Cerebral Arteries/injuries , Vascular Patency/physiology , Animals , Brain/blood supply , Brain Ischemia/physiopathology , Capillary Resistance/physiology , Corpus Striatum/blood supply , Corpus Striatum/pathology , Disease Models, Animal , Evans Blue/analysis , Evans Blue/metabolism , Fluorescent Dyes , Glucose/administration & dosage , Glucose/pharmacology , Hyperglycemia , Infusions, Intravenous , Male , Microcirculation/physiology , Neocortex/blood supply , Neocortex/pathology , Rats , Rats, Wistar , Reperfusion Injury/physiopathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...