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










Publication year range
1.
Dev Neurosci ; 23(3): 192-7, 2001.
Article in English | MEDLINE | ID: mdl-11598319

ABSTRACT

The aim of this study was to evaluate the involvement of mitochondrial membrane permeability transition (MPT) after hypoxia-ischemia (HI) in 7-day-old rats. [14C]2-deoxyglucose (DOG) was administered to controls, and at various time points after HI. MPT in the cerebral cortex was measured as entrapment of DOG-6-P in mitochondria. Another group of rats was treated with the MPT inhibitor cyclosporin A (CsA; 10-50 mg/kg i.p.) or vehicle before and after HI, and the effect on brain injury and mitochondrial respiration was evaluated. A significant increase in DOG-6-P entrapment in mitochondria indicated that MPT occurred in two phases: a primary MPT after 0-1.5 h and a secondary MPT after 6.5-8 h of reperfusion. However, CsA did not affect brain injury or mitochondrial respiration. The data suggest that MPT occurred after HI but does not provide evidence for its involvement in the development of injury.


Subject(s)
Glucose-6-Phosphate/analogs & derivatives , Hypoxia-Ischemia, Brain/metabolism , Mitochondria/metabolism , Animals , Animals, Newborn , Brain/metabolism , Carbon Radioisotopes , Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Glucose-6-Phosphate/pharmacokinetics , Rats
2.
Neuroreport ; 11(13): 2833-6, 2000 Sep 11.
Article in English | MEDLINE | ID: mdl-11006950

ABSTRACT

The aim was to study the effects of an NMDA receptor antagonist on caspase-3 activation and DNA fragmentation after hypoxia-ischemia (HI) in 7-day-old rats. Animals were treated with vehicle or MK-801 (0.5 mg/kg) directly after HI and sacrificed 8, 24 or 72h later. MK-801 reduced injury (by 53%), cells positive for active caspase-3 (by 39%) and DNA fragmentation (by 79%) in the cerebral cortex. Furthermore, MK-801 significantly decreased caspase-3 activity, and Western blots revealed a tendency towards decreased proteolytic cleavage of the caspase-3 proform. The data imply that NMDA receptors are involved in the activation of apoptotic processes in the immature brain after HI.


Subject(s)
Asphyxia Neonatorum/metabolism , Caspases/metabolism , DNA Fragmentation/drug effects , Dizocilpine Maleate/pharmacology , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/physiopathology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Animals, Newborn/injuries , Animals, Newborn/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Asphyxia Neonatorum/drug therapy , Asphyxia Neonatorum/physiopathology , Caspase 3 , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , DNA Fragmentation/physiology , Female , Humans , Hypoxia-Ischemia, Brain/drug therapy , Infant, Newborn , Male , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism
3.
Neurochem Int ; 37(2-3): 307-16, 2000.
Article in English | MEDLINE | ID: mdl-10812216

ABSTRACT

This in vivo study, aimed at detecting the N-methyl-D-aspartate (NMDA) evoked Ca(2+)-induced Ca(2+) release from intracellular stores in the neonatal rat brain, demonstrates that the application of 5 mM N-methyl-D-aspartate via a microdialysis probe for 20 min to the dentate gyrus (DG) of halotane-anesthetized 7 day-old (postnatal day 7, PND 7) rats induces a prolonged decrease in Ca(2+) concentration in an initially calcium-free dialysis medium, indicative of a drop in the extracellular concentration of Ca(2+) and Ca(2+) influx to neurons. In parallel experiments, a huge NMDA-evoked release of 45Ca from the pre-labeled endogenous Ca(2+) pool was observed and interpreted as the expression of intracellular Ca(2+) release. Dantrolene (100 microM) significantly inhibited the NMDA-induced 45Ca release, whereas 250 microM ryanodine exerted an unspecific biphasic effect. Autoradiographic and immunocytochemical detection of ryanodine receptors and calbindin D(28K), respectively, in the hippocampal region of PND 7 rats displayed a pronounced expression of [3H]ryanodine binding sites in the DG, but only a slight immunoreactivity of calbindin D(28K). Plastic changes in neurons or excitotoxic neuronal damage induced by the activation of NMDA receptors are mediated by Ca(2+) signals, resulting from an influx of extracellular Ca(2+), and also in some neurons, from the release of intracellular Ca(2+). Our previous in vivo microdialysis experiments visualized NMDA-evoked 45Ca release in the adult rat dentate gyrus, attributable to Ca(2+)-induced Ca(2+) release from the ryanodine-sensitive pool. An additional role of calbindin in the mechanism of this phenomenon has been suggested. This aspect has not been studied in vivo in newborn rats. Our present results indicate that the release of 45Ca from the prelabeled intracellular, dantrolene-sensitive Ca(2+) pool in the DG neurons of immature rats, most probably representing a phenomenon of Ca(2+)-induced Ca(2+) release, significantly participates in the generation of NMDA receptor-mediated intracellular Ca(2+) signals, whereas the role of calbindin D(28K) in the mechanism of 45Ca release is negligible.


Subject(s)
Animals, Newborn/metabolism , Calcium/metabolism , Dentate Gyrus/metabolism , Excitatory Amino Acid Agonists/pharmacology , N-Methylaspartate/pharmacology , Animals , Autoradiography , Calbindins , Calcium Radioisotopes , Dantrolene/pharmacology , Dentate Gyrus/drug effects , Immunohistochemistry , Microdialysis , Muscle Relaxants, Central/pharmacology , Rats , Rats, Wistar , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , S100 Calcium Binding Protein G/metabolism
4.
Brain Res Dev Brain Res ; 125(1-2): 31-41, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11154758

ABSTRACT

Recent data imply that mitochondrial regulation of calcium is critical in the process leading to hypoxic-ischemic brain injury. The aim was to study the subcellular distribution of calcium in correlation with ultrastructural changes after hypoxia-ischemia in neonatal rats. Seven-day-old rats were subjected to permanent unilateral carotid artery ligation and exposure to hypoxia (7.7% oxygen in nitrogen) for 90 min. Animals were perfusion-fixed after 30 min, 3 h or 24 h of reperfusion. Sections were sampled for light microscopy and electron microscopy combined with the oxalate-pyroantimonate technique. At 30 min and 3 h of reflow, a progressive accumulation of calcium was detected in the endoplasmic reticulum, cytoplasm, nucleus and, most markedly, in the mitochondrial matrix of neurons in the gray matter in the core area of injury. Some mitochondria developed a considerable degree of swelling reaching a diameter of several microm at 3 h of reflow whereas the majority of mitochondria appeared moderately affected. Chromatin condensation was observed in nuclei of many cells with severely swollen mitochondria with calcium deposits. A whole spectrum of morphological features ranging from necrosis to apoptosis was seen in degenerating cells. After 24 h, there was extensive injury in the cerebral cortex as judged by breaks of mitochondrial and plasma membranes, and a general decrease of cellular electron density. In the white matter of the core area of injury, the axonal elements exhibited varicosity-like swellings filled with calcium-pyroantimonate deposits. Furthermore, the thin myelin sheaths were loaded with calcium. Numerous oligodendroglia-like cells displayed apoptotic morphology with shrunken cytoplasm and chromatin condensation, whereas astroglial necrosis was not seen. In conclusion, markedly swollen 'giant' mitochondria with large amounts of calcium were found at 3 h of reperfusion often in neuronal cells with condensation of the nuclear chromatin. The results are discussed in relation to mitochondrial permeability transition and activation of apoptotic processes.


Subject(s)
Calcium/analysis , Hypoxia-Ischemia, Brain/pathology , Neurons/chemistry , Animals , Animals, Newborn , Antimony , Apoptosis , Cerebral Cortex/chemistry , Cerebral Cortex/cytology , Female , Male , Microscopy, Electron , Mitochondria/chemistry , Mitochondria/pathology , Mitochondria/ultrastructure , Mitochondrial Swelling , Nerve Degeneration/pathology , Nerve Fibers/chemistry , Nerve Fibers/ultrastructure , Neurons/ultrastructure , Oxalates , Rats , Rats, Wistar , Reperfusion Injury/pathology
5.
Brain Res Dev Brain Res ; 125(1-2): 43-50, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11154759

ABSTRACT

Mitochondrial damage may play a key role in the development of necrotic and apoptotic hypoxic-ischemic (HI) brain damage. It has previously been shown that mitochondrial respiration is depressed in the cerebral cortex after HI in neonatal animals. The aim of the present study was to further characterize the time course of the mitochondrial impairment during reperfusion and the correlation between the respiratory control ratio and brain injury and activation of caspase-3. Rat pups were subjected to unilateral carotid artery ligation and exposed to hypoxia (7.7% oxygen). Mitochondrial respiration was measured 0-72 h after HI in a mitochondrial fraction isolated from cerebral cortex. Microtubule associated protein-2 (MAP2) and caspase-3 were analyzed with immunoblotting in cerebral cortex homogenates. In addition, the time course of caspase-3 activation was measured as DEVD cleavage. The mitochondrial respiratory control ratio in cerebral cortex decreased immediately after HI followed by a partial recovery at 3-8 h. Thereafter, a secondary drop occurred with a minimum reached at 24 h of reperfusion. The secondary loss of respiratory function was accompanied by depletion of MAP2, cleavage of caspase-3 and an increased caspase-3 -like activity at 3-24 h after the insult. In conclusion, the primary phase of mitochondrial dysfunction was paralleled by a moderate decrease of MAP2 and a limited activation of caspase-3. The secondary mitochondrial impairment was associated with neuronal injury and pronounced activation of caspase-3.


Subject(s)
Caspases/metabolism , Hypoxia-Ischemia, Brain/metabolism , Mitochondria/metabolism , Neurons/enzymology , Animals , Animals, Newborn , Antibody Specificity , Blotting, Western , Caspase 3 , Cell Respiration/physiology , Cerebral Cortex/pathology , Enzyme Activation/physiology , Female , Hypoxia-Ischemia, Brain/pathology , Male , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/immunology , Neurons/chemistry , Neurons/pathology , Rats , Rats, Inbred WF
6.
Brain Res Dev Brain Res ; 125(1-2): 51-60, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11154760

ABSTRACT

Hypoxic-ischemic brain injury involves an increased formation of reactive oxygen species. Key factors in the cellular protection against such agents are the GSH-associated reactions. In the present study we examined alterations in total glutathione and GSSG concentrations in mitochondria-enriched fractions and tissue homogenates from the cerebral cortex of 7-day-old rats at 0, 1, 3, 8, 14, 24 and 72 h after hypoxia-ischemia. The concentration of total glutathione was transiently decreased immediately after hypoxia-ischemia in the mitochondrial fraction, but not in the tissue, recovered, and then decreased both in mitochondrial fraction and homogenate after 14 h, reaching a minimum at 24 h after hypoxia-ischemia. The level of GSSG was approximately 4% of total glutathione and increased selectively in the mitochondrial fraction immediately after hypoxia-ischemia. The decrease in glutathione may be important in the development of cell death via impaired free radical inactivation and/or redox related changes. The effects of hypoxia-ischemia on the concentrations of selected amino acids varied. The levels of phosphoethanolamine, an amine previously reported to be released in ischemia, mirrored the changes in glutathione. GABA concentrations initially increased (0-3 h) followed by a decrease at 72 h. Glutamine levels increased, whereas glutamate and aspartate were unchanged up to 24 h after the insult. The results on total glutathione and GSSG are discussed in relation to changes in mitochondrial respiration and microtubule associated protein-2 (MAP2) which are reported on in accompanying paper [64].


Subject(s)
Amino Acids/metabolism , Cerebral Cortex/metabolism , Glutathione/metabolism , Hypoxia-Ischemia, Brain/metabolism , Age Factors , Animals , Carotid Artery, Common , Cell Respiration/physiology , Cerebral Cortex/chemistry , Cerebral Cortex/growth & development , Ethanolamines/metabolism , Female , Glutathione Disulfide/metabolism , Ligation , Male , Microtubule-Associated Proteins/analysis , Mitochondria/metabolism , Oxidative Stress/physiology , Rats , Rats, Inbred WF
7.
J Biol Chem ; 274(20): 14046-52, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10318818

ABSTRACT

In a model of cerebral hypoxia-ischemia in the immature rat, widespread brain injury is produced in the ipsilateral hemisphere, whereas the contralateral hemisphere is left undamaged. Previously, we found that calpains were equally translocated to cellular membranes (a prerequisite for protease activation) in the ipsilateral and contralateral hemispheres. However, activation, as judged by degradation of fodrin, occurred only in the ipsilateral hemisphere. In this study we demonstrate that calpastatin, the specific, endogenous inhibitor protein to calpain, is up-regulated in response to hypoxia and may be responsible for the halted calpain activation in the contralateral hemisphere. Concomitantly, extensive degradation of calpastatin occurred in the ipsilateral hemisphere, as demonstrated by the appearance of a membrane-bound 50-kDa calpastatin breakdown product. The calpastatin breakdown product accumulated in the synaptosomal fraction, displaying a peak 24 h post-insult, but was not detectable in the cytosolic fraction. The degradation of calpastatin was blocked by administration of CX295, a calpain inhibitor, indicating that calpastatin acts as a suicide substrate to calpain during hypoxia-ischemia. In summary, calpastatin was up-regulated in areas that remain undamaged and degraded in areas where excessive activation of calpains and infarction occurs.


Subject(s)
Brain Ischemia/metabolism , Calcium-Binding Proteins/metabolism , Calpain/metabolism , Hypoxia/metabolism , Up-Regulation , Animals , Animals, Newborn , Brain/drug effects , Brain/enzymology , Brain/pathology , Cell Membrane/enzymology , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Enzyme Activation , Female , Functional Laterality , Male , Rats , Rats, Wistar
8.
Pediatr Res ; 45(4 Pt 1): 500-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10203141

ABSTRACT

Hypoxia-ischemia induces an inflammatory response in the immature central nervous system that may be important for development of brain injury. Recent data implicate that chemoattractant cytokines, chemokines, are involved in the recruitment of immune cells. The aim was to study alpha- and beta-chemokines in relation to the temporal activation of inflammatory cells after hypoxia-ischemia in immature rats. Hypoxia-ischemia was induced in 7-day-old rats (left carotid artery occlusion + 7.7% oxygen). The pups were decapitated at different times after the insult. Immunohistochemistry was used for evaluation of the inflammatory cell response and RT-PCR to analyze the cytokine mRNA and chemokine mRNA expression. A distinct interleukin-1beta and tumor necrosis factor-alpha cytokine expression was found 0-24 h after hypoxia-ischemia that was accompanied by induction of alpha-chemokines (growth related gene and macrophage inflammatory protein-2). In the next phase, the beta2-integrin expression was increased (12 h and onward) and neutrophils transiently invaded the vessels and tissue in the infarct region. The mRNA induction for the beta-chemokines macrophage inflammatory protein-1alpha, macrophage inflammatory protein-1beta, and RANTES preceded the expression of markers for lymphocytes [cluster of differentiation (CD)4, CD8], microglia/macrophages (MHC I), and natural killer cells in the infarct area. The activation of microglia/macrophages, CD4 lymphocytes, and astroglia persisted up to at least 42 d of postnatal age implicating a chronic component of immunoinflammatory activation. The expression of mRNA for alpha- and beta-chemokines preceded the appearance of immune cells suggesting that these molecules may have a role in the inflammatory response to insults in the immature central nervous system.


Subject(s)
Brain Ischemia/immunology , Brain/immunology , Cerebral Infarction/immunology , Chemokines/genetics , Cytokines/genetics , Hypoxia, Brain/immunology , Animals , Animals, Newborn , Brain/pathology , Brain Ischemia/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cerebral Infarction/genetics , Chemokine CCL5/genetics , Female , Gene Expression Regulation , Hypoxia, Brain/genetics , Ischemic Attack, Transient/genetics , Ischemic Attack, Transient/immunology , Lymphocyte Activation , Macrophages/immunology , Male , Microglia/immunology , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
9.
Pediatr Res ; 44(5): 746-54, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9803457

ABSTRACT

Cerebral lactate rises after chemically induced seizures, but it is not known if this occurs with posthypoxic seizures. We examined changes in lactate and pyruvate in gray and white matter in the newborn pig brain after a hypoxic insult known to produce seizures and permanent brain damage. Fourteen halothane-anesthetized piglets aged 24-49 h, were instrumented with a two-channel scalp EEG and microdialysis probes positioned in white and gray matter. Forty-five minutes of hypoxia were induced by reducing the fraction of inspired O2 to the maximum concentration at which EEG amplitude was < 7 microV. Postinsult EEG was classified as electroconvulsive activity (ECA) (n = 4) or burst suppression (n = 2), persistently low amplitude (n = 2), or intermittent spikes on normal background activity (n = 6). Six hours after the insult the brains were perfusion fixed for histologic probe localization. Plasma lactate and brain lactate had different time courses with brain having a persistently elevated lactate/pyruvate (L/P) ratio. The highest L/P ratios in gray and white matter were in the two pigs with persistently low amplitude EEG. There was no association between onset of electroconvulsive activity and an increase in lactate or L/P ratio. Posthypoxic energy metabolism is disturbed in both gray and white matter probably because of mitochondrial dysfunction. Seizure activity does not increase cerebral lactate or L/P ratio above the already raised levels found in posthypoxic encephalopathy. These findings cast further doubt on the hypothesis that such seizures are, in themselves, damaging.


Subject(s)
Cerebellum/metabolism , Hypoxia/complications , Lactic Acid/metabolism , Pyruvic Acid/metabolism , Seizures/metabolism , Animals , Animals, Newborn , Electroencephalography , Microdialysis , Seizures/etiology , Swine , Time Factors
10.
Brain Res ; 797(2): 328-32, 1998 Jun 29.
Article in English | MEDLINE | ID: mdl-9666161

ABSTRACT

The aim of this study was to investigate the role of cysteine in development of brain damage after hypoxia-ischemia (HI) in neonatal rats. Rat pups were subjected to unilateral carotid ligation and exposure to hypoxia (7.7% oxygen) for 60 or 90 min. A subtoxic dose of cysteine were administered before or after HI and the unilateral brain injury was evaluated 14 days after the insult and expressed as ipsilateral weight deficit as % of the contralateral hemisphere. In some experiments the changes of extracellular (e.c.) cysteine in the cerebral cortex were sampled with microdialysis and analyzed with HPLC. Cysteine in a dose of 0.2 mg/g s.c. given before 60 min of HI increased the extent of brain injury by 59%. The effect of posttreatment was limited and dependent on the duration of HI: 0.2 mg/g of cysteine given after 90 min of HI increased the degree of brain injury by 25%, whereas the same dose administered after 60 min of HI was ineffective in spite of that this combination of cysteine and HI resulted in e.c. cysteine concentrations 3-4 times higher than those observed in non-treated HI controls. These data show that subtoxic doses of cysteine administered before or after HI enhances brain injury. However, e.c. cysteine levels exceeding those induced by HI are required which makes a substantial contribution of cysteine in the pathophysiology of HI brain injury in the neonatal rat unlikely.


Subject(s)
Brain Chemistry/drug effects , Brain Ischemia/physiopathology , Cysteine/pharmacology , Hypoxia, Brain/physiopathology , Animals , Animals, Newborn , Female , Male , Microdialysis , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Rats , Rats, Wistar , Sulfur/physiology
11.
J Cereb Blood Flow Metab ; 18(3): 297-304, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9498846

ABSTRACT

Treatment after hypoxia-ischemia (HI) in immature rats with the N-methyl-D-aspartate receptor (NMDAR) antagonist dizocilpine maleate (MK-801) reduces areas with high glucose utilization and reduces brain damage. The object was to study the metabolic effects of MK-801 treatment after HI. Seven-day-old rats were randomized to the following groups: non-HI, HI, or HI plus MK-801 (0.5 mg/kg immediately after HI). In the parietal cortex, the mitochondrial respiration was measured in homogenates 1 to 4 hours, and the energy metabolites at 3 and 8 hours after HI. The energy use was calculated from changes in energy metabolites after decapitation at 3 hours after HI. State 3 respiration was reduced by 46%, 32%, and 25% after HI compared with non-HI with pyruvate plus malate, glutamate plus malate, or glutamate plus succinate as substrates, respectively. Uncoupler-stimulated but not state 4 respiration was similarly reduced. The MK-801 augmented pyruvate plus malate-supported state 3 respiration after HI by 42%. The energy utilization was not affected by HI but was reduced by MK-801 treatment in the ipsilateral cortex from 4.6 +/- 2.3 to 2.6 +/- 1.8 micromol high-energy phosphate bond/min/g. The levels of ATP and phosphocreatine did not differ between the HI and HI plus MK-801 groups at 3 hours, but were lower in the HI than in the HI plus MK-801 group at 8 hours after HI. In conclusion, treatment with MK-801 reduced energy utilization and improved mitochondrial function and energy status after HI, suggesting a linkage between NMDAR activation and impaired energy metabolism during reperfusion.


Subject(s)
Brain Ischemia/metabolism , Brain/blood supply , Brain/metabolism , Energy Metabolism , Mitochondria/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Brain/growth & development , Brain/ultrastructure , Brain Ischemia/pathology , Mitochondria/pathology , Rats , Rats, Wistar
12.
Neuroreport ; 8(15): 3359-62, 1997 Oct 20.
Article in English | MEDLINE | ID: mdl-9351672

ABSTRACT

Hypothermia applied after hypoxia offers neuroprotection in neonatal animals, but the mechanisms involved remain unknown. Hypoxia was induced in newborn piglets and changes in excitatory amino acids (EAAs) and the citrulline:arginine ratio (CAR) were followed by microdialysis for 5 h. After the 45 min hypoxic insult, the animals were randomized to receive normothermia (39 degrees C; n=7) or hypothermia (35 degrees C; n = 7). After reoxygenation, extracellular glutamate, aspartate and the excitotoxic index were significantly lower in the cerebral cortex of hypothermic animals than in normothermic animals. A progressive rise of the CAR occurred during reoxygenation in the normothermic group whereas the ratio tended to decrease in the hypothermic group. In conclusion, post-hypoxic hypothermia attenuated NO production and overflow of EAAs.


Subject(s)
Cerebral Cortex/metabolism , Excitatory Amino Acids/metabolism , Hypothermia/metabolism , Hypoxia, Brain/metabolism , Nitric Oxide/metabolism , Animals , Arginine/metabolism , Citrulline/metabolism , Electroencephalography , Microdialysis , Swine
13.
Acta Paediatr Suppl ; 422: 85-8, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9298801

ABSTRACT

The Levene model in 7-day-old rats is the most often used model of hypoxia-ischaemia (HI) in immature animals. The rat central nervous system is immature at birth and corresponds neurodevelopmentally to the term human infant during the second postnatal week. The Levene model of HI differs from clinical asphyxia with respect to the unilateral distribution of brain injury and lack of multi-organ dysfunction. Furthermore, it does not allow cardiovascular monitoring or repeated blood sampling. On the other hand, the progressive nature of HI bears many similarities to birth asphyxia with regard to blood flow changes and cellular metabolic derangements. The model is well characterized, easy to carry out and the low cost allows inclusion of a sufficient number of animals for dose-response evaluation of neuroprotective agents. In addition, it provides the unique opportunity of long-term evaluation of neuropathological and functional outcome.


Subject(s)
Brain Ischemia/physiopathology , Disease Models, Animal , Hypoxia/physiopathology , Animals , Animals, Newborn , Asphyxia Neonatorum , Brain Ischemia/metabolism , Humans , Hypoxia/metabolism , Infant, Newborn , Rats , Species Specificity
14.
Brain Res ; 750(1-2): 325-8, 1997 Mar 07.
Article in English | MEDLINE | ID: mdl-9098561

ABSTRACT

The aim of this study was to follow extracellular concentrations of excitatory amino acids (EAAs) and cysteine during neonatal hypoxia-ischemia (HI) and reflow and to relate these events to the extent of brain damage evaluated 6 h after the insult. Rat pups (PND 7-10) were subjected to unilateral ligation of the common carotid artery and exposed to hypoxia (7.7% O2). Extracellular amino acids were sampled during HI and for 6 h of reperfusion with microdialysis and the levels were correlated with the extent of brain damage at the site of probe placement. The concentrations of glutamate, aspartate and cysteine increased transiently during HI (15 x, 6 x and 3 x, respectively) in the extracellular space and returned to normal or remained slightly elevated during reperfusion. Changes of EAAs and cysteine were similar during HI in the infarcted, undamaged and border-zone regions. During reperfusion the concentrations of glutamate, aspartate and cysteine were higher in infarcted and border-zone areas compared to undamaged tissue. In neonatal rats, the extracellular levels of EAAs during HI do not correspond to the extent of brain injury whereas the EAA concentrations during reflow are related to the extent of infarction.


Subject(s)
Brain Injuries/physiopathology , Brain/metabolism , Cysteine/metabolism , Excitatory Amino Acids/metabolism , Hypoxia, Brain/physiopathology , Ischemic Attack, Transient/physiopathology , Analysis of Variance , Animals , Animals, Newborn , Aspartic Acid/metabolism , Brain Injuries/etiology , Carotid Artery, Common , Extracellular Space , Factor Analysis, Statistical , Female , Glutamic Acid/metabolism , Male , Microdialysis , Rats , Rats, Wistar , Reperfusion , Statistics, Nonparametric
15.
Metab Brain Dis ; 11(2): 109-23, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8776714

ABSTRACT

The aim of this study was to investigate the possible role of excitatory amino acids (EAAs) and cysteine in the development of brain damage after hypoxia-ischemia (HI) in neonates. In a rat model of neonatal HI, changes in extracellular (ec) amino acids in cerebral cortex were measured with microdialysis and correlated with the extent of brain damage at the site of probe placement. Extracellular concentrations of glutamate, aspartate and cysteine increased during HI and remained elevated during reperfusion. During HI the pattern of EAA changes was the same in the infarcted, undamaged and border zone regions. During reperfusion, however, the ec concentrations of glutamate, aspartate and cysteine were higher in infarcted and border zone areas compared to undamaged tissue. HI also produced a slight increase of tissue concentration of cysteine and decrease of tissue concentration of glutamate in parietal cortex of the HI hemisphere. The effect of cysteine on brain damage induced by HI and glutamate was also investigated. A subtoxic dose of cysteine potentiated glutamate toxicity in the arcuate nucleus and enhanced brain infarction after HI in neonatal rats. The results show that in neonatal HI the extracellular levels of EAAs during HI are not directly related to brain injury but the EAA levels during reflow predict the extent of infarction. Cysteine increases HI-induced brain injury and potentiates glutamate toxicity in neonatal rats. Speculatively, elevated level of cysteine during reperfusion may participate in the excitotoxic cascade leading to brain injury.


Subject(s)
Brain Diseases/chemically induced , Brain Ischemia/chemically induced , Cysteine/pharmacology , Excitatory Amino Acids/pharmacology , Animals , Glutamic Acid/pharmacology , Rats , Time Factors
16.
Brain Res ; 705(1-2): 65-70, 1995 Dec 24.
Article in English | MEDLINE | ID: mdl-8821734

ABSTRACT

L-Cysteine produces excitotoxic brain damage but its chemical structure differs from that of other excitotoxins. Although it is an NMDAmimetic, its mode of action is complex and may encompass antiexcitotoxic components. The purpose of the present study was to investigate whether cysteine kills neurons by potentiating the effects of glutamate and/or by releasing glutamate. In primary cultures of cortical neurons, 24 h of exposure to glutamate caused a concentration-dependent, dizocilpine-sensitive cell death as measured by release of lactate dehydrogenase. Cysteine was also toxic but higher concentrations were required. In addition, N-acetylcysteine produced mild toxicity at 1 mM. There was no general potentiation between either glutamate and cysteine or glutamate and N-acetylcysteine although some combinations acted synergistically. In no case did the thiols inhibit glutamate toxicity. The interaction between glutamate and cysteine toxicity was also assessed in the immature rat arcuate nucleus in vivo. When given at a dose (0.5 mg/g) that did not cause any toxicity per se, cysteine enhanced the toxicity of glutamate (0.3-0.8 mg/g). Cortical microdialysis was carried out in anesthetized rats (8-10 days old) administered a toxic dose of cysteine (1 mg/g). The levels of taurine were elevated 15-fold, phosphoethanolamine 3-fold and alanine 2-fold. Despite the observation that glutamine decreased markedly and rapidly, there was only a delayed doubling of glutamate concentrations. It is therefore unlikely that cysteine induces neurotoxicity by releasing glutamate. Taken together, the results suggest that there is a synergistic effect between cysteine and glutamate. Speculatively, this potentiation may be produced by reduction by cysteine of the redox site of the glutamate-activated NMDA receptor-ionophore complex.


Subject(s)
Cysteine/toxicity , Glutamic Acid/drug effects , Neurotoxins/pharmacology , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Dizocilpine Maleate/pharmacology , Ethanolamines/metabolism , Glutamic Acid/analysis , Glutamic Acid/toxicity , Microdialysis , Neurons/cytology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Taurine/metabolism
17.
Int J Dev Neurosci ; 13(7): 685-704, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8787860

ABSTRACT

This in vivo study concerns developmental differences in the sensitivity of striatal neurons to N-methyl-D-aspartate (NMDA). Changes in calcium homeostasis in adult vs immature rats at postnatal days 8-10, evoked by NMDA, were evaluated by measurements of 45Ca efflux and of Ca2+ taurine and phosphoethanolamine concentrations in striatal microdialysates. The efflux of [14C]sucrose was employed in order to measure changes in extracellular space volume. In adult rats the addition of 5 mM NMDA for 20 min to the perfusion medium resulted in a 30-40% increase in 45Ca efflux, and in a 15% increase in [14C]sucrose efflux. Ten minutes after NMDA perfusion, 45Ca and [14C] sucrose efflux returned to the baseline. No significant changes in Ca2+ or amino acid concentrations were observed in the dialysate of the adult rat striatum. NMDA perfusion in the striatum of immature rats initially induced a transient (5 min) increase in the efflux of 45Ca (by 13%) and [14C]sucrose (by 9%). This was followed by a prolonged (lasting 45-50 min) 45% decrease in 45Ca efflux, an 80% reduction of Ca2+ concentration, and increases in taurine and phosphoethanolamine concentrations in the dialysate, whereas [14C]sucrose efflux recovered within 10 min. These data illustrate differences in the NMDA response between developing and adult rat striatum. Only in developing rats did NMDA induce a large and prolonged influx of extracellular calcium to neurons that may explain the enhanced NMDA neurotoxicity in immature rats.


Subject(s)
Calcium/physiology , Excitatory Amino Acid Agonists/pharmacology , Homeostasis/drug effects , N-Methylaspartate/pharmacology , Neostriatum/drug effects , Neostriatum/growth & development , Aging/physiology , Amino Acids/metabolism , Animals , Calcium Radioisotopes , Microdialysis , Neostriatum/anatomy & histology , Rats , Rats, Wistar , Sucrose/metabolism
18.
Acta Neurobiol Exp (Wars) ; 55(4): 223-31, 1995.
Article in English | MEDLINE | ID: mdl-8713352

ABSTRACT

We report the results of microdialysis experiments investigating the NMDA-induced release of intracellular Ca2+ in different brain regions. Microdialysis probes were implanted stereotaxically into the striatum, thalamus and hippocampus dentate gyrus (DG) of adult rats. Dialysates were analysed for alterations in the concentration of ionized Ca2+ in an initially calcium-free medium and for changes in 45Ca efflux from the pre-labelled endogenous Ca2+ pools. The application of 5 mM of NMDA to the dialysis medium for 20 min in the striatum, resulted in increases in Ca2+ and 45Ca concentrations by 25% and 35% respectively. After NMDA perfusion in the hippocampus DG and in the thalamus, decreases in the Ca2+ concentration to 65.6% and 38.6% of the basal level respectively, were accompanied by increases in 45Ca efflux, exceeding 1,500% of the basal level in the hippocampus. Cell swelling, and the corresponding reduction of the extracellular space volume was insufficient to explain the huge increase in 45Ca efflux. Thus, our experiments demonstrated that in vivo in the rat hippocampus DG, NMDA induces the release of 45Ca to the extracellular space from unidentified intracellular calcium stores.


Subject(s)
Calcium/metabolism , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/metabolism , N-Methylaspartate/pharmacology , Animals , Calcium Radioisotopes , Hippocampus/drug effects , Microdialysis , Rats , Rats, Wistar
19.
Brain Res Dev Brain Res ; 83(1): 79-84, 1994 Nov 18.
Article in English | MEDLINE | ID: mdl-7697873

ABSTRACT

In a model of perinatal hypoxia-ischemia (HI) we examined the neuroprotective efficacy of pre- and post-treatment with the glutamate release inhibitor BW1003C87 [5-(2,3,5-trichlorophenyl)-2,4-diamino-pyrimidine). Ipsilateral brain damage developed in 99% of rat pups subjected to HI (unilateral common carotid artery ligation and 100 min of 7.7% oxygen exposure) with a 26 +/- 16% (mean +/- S.D.) weight deficit of the damaged hemisphere 2 weeks after the insult. Pre-treatment with BW1003C87 (10 mg/kg intraperitoneally) reduced the brain damage by 46% (P < 0.05). A higher dose (20 mg/kg) of pre-treatment was not tolerated. Administration of BW1003C87 did not affect the rectal temperature of the rats. Post-treatment with BW1003C87 (10-30 mg/kg) offered no neuroprotection in this model. In conclusion, there was a neuroprotective effect from pre- but not post-treatment with BW1003C87 in this model, supporting the concept that intra-ischemic excitatory amino acid release is important for development of brain damage. The lack of post-treatment effect indicates that BW1003C87 did not attenuate deleterious EAA cycling during reflow in the neonatal brain.


Subject(s)
Brain/pathology , Excitatory Amino Acid Antagonists , Hypoxia, Brain/prevention & control , Ischemic Attack, Transient/prevention & control , Pyrimidines/pharmacology , Animals , Animals, Newborn , Brain/anatomy & histology , Brain Damage, Chronic/pathology , Cerebral Infarction/pathology , Cerebral Infarction/prevention & control , Female , Functional Laterality , Hypoxia, Brain/pathology , Ischemic Attack, Transient/pathology , Male , Organ Size , Premedication , Pyrimidines/toxicity , Rats , Rats, Inbred WF
20.
Brain Res Dev Brain Res ; 77(1): 146-50, 1994 Jan 14.
Article in English | MEDLINE | ID: mdl-8131260

ABSTRACT

The extracellular calcium concentration ([Ca2+]ec) was recorded by calcium-sensitive microelectrodes in the parietal cortex of 9-11 day old rats during anoxia. During the first 10 min of anoxia, [Ca2+]ec increased from 1.1 mM to 1.5 +/- 0.23 mM, and thereafter it started to decrease reaching below basal level after around 13 min. The [Ca2+]ec decrease was either slow and continuous, or biphased with a rapid initial decrease followed by a continuous slow decrease. After 60 min of anoxia, the [Ca2+]ec had reached 0.2-0.3 mM. Changes in [Ca2+]ec in animals treated with the NMDA receptor antagonist MK-801 (0.3 mg/kg i.p.) did not display any significant differences compared to controls. Thus, the strong neuroprotective effect of MK-801 in ischemic situations in the immature brain can not be explained by a prevention of calcium entry during anoxic depolarization.


Subject(s)
Calcium/metabolism , Cerebral Cortex/metabolism , Dizocilpine Maleate/pharmacology , Extracellular Space/metabolism , Hypoxia/metabolism , Animals , Animals, Newborn , Osmolar Concentration , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...