Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Mol Neurobiol ; 60(4): 2024-2035, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36598650

ABSTRACT

The present study aimed to elucidate the effect of sulforaphane (a natural isothiocyanate) on oxidative stress and mitochondrial dysfunction during and at selected periods following status epilepticus (SE) induced in immature 12-day-old rats by Li-pilocarpine. Dihydroethidium was employed for the detection of superoxide anions, immunoblot analyses for 3-nitrotyrosine (3-NT) and 4-hydroxynonenal (4-HNE) levels and respiratory chain complex I activity for evaluation of mitochondrial function. Sulforaphane was given i.p. in two doses (5 mg/kg each), at PD 10 and PD 11, respectively. The findings of the present study indicate that both the acute phase of SE and the early period of epileptogenesis (1 week and 3 weeks following SE induction) are associated with oxidative stress (documented by the enhanced superoxide anion production and the increased levels of 3-NT and 4-HNE) and the persisting deficiency of complex I activity. Pretreatment with sulforaphane either completely prevented or significantly reduced markers of both oxidative stress and mitochondrial dysfunction. Since sulforaphane had no direct anti-seizure effect, the findings suggest that the ability of sulforaphane to activate Nrf2 is most likely responsible for the observed protective effect. Nrf2-ARE signaling pathway can be considered a promising target for novel therapies of epilepsy, particularly when new compounds, possessing inhibitory activity against protein-protein interaction between Nrf2 and its repressor protein Keap1, with less "off-target" effects and, importantly, with an optimal permeability and bioavailability properties, become available commercially.


Subject(s)
NF-E2-Related Factor 2 , Status Epilepticus , Rats , Animals , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Isothiocyanates/pharmacology , Sulfoxides/metabolism , Sulfoxides/pharmacology , Status Epilepticus/metabolism , Superoxides/metabolism , Mitochondria/metabolism
2.
Front Cell Neurosci ; 16: 855161, 2022.
Article in English | MEDLINE | ID: mdl-35370554

ABSTRACT

Status epilepticus (SE) is a common paediatric emergency with the highest incidence in the neonatal period and is a well-known epileptogenic insult. As previously established in various experimental and human studies, SE induces long-term alterations to brain metabolism, alterations that directly contribute to the development of epilepsy. To influence these changes, organic isothiocyanate compound sulforaphane (SFN) has been used in the present study for its known effect of enhancing antioxidative, cytoprotective, and metabolic cellular properties via the Nrf2 pathway. We have explored the effect of SFN in a model of acquired epilepsy induced by Li-Cl pilocarpine in immature rats (12 days old). Energy metabolites PCr, ATP, glucose, glycogen, and lactate were determined by enzymatic fluorimetric methods during the acute phase of SE. Protein expression was evaluated by Western blot (WB) analysis. Neuronal death was scored on the FluoroJadeB stained brain sections harvested 24 h after SE. To assess the effect of SFN on glucose metabolism we have performed a series of 18F-DG µCT/PET recordings 1 h, 1 day, and 3 weeks after the induction of SE. Responses of cerebral blood flow (CBF) to electrical stimulation and their influence by SFN were evaluated by laser Doppler flowmetry (LDF). We have demonstrated that the Nrf2 pathway is upregulated in the CNS of immature rats after SFN treatment. In the animals that had undergone SE, SFN was responsible for lowering glucose uptake in most regions 1 h after the induction of SE. Moreover, SFN partially reversed hypometabolism observed after 24 h and achieved full reversal at approximately 3 weeks after SE. Since no difference in cell death was observed in SFN treated group, these changes cannot be attributed to differences in neurodegeneration. SFN per se did not affect the glucose uptake at any given time point suggesting that SFN improves endogenous CNS ability to adapt to the epileptogenic insult. Furthermore, we had discovered that SFN improves blood flow and accelerates CBF response to electrical stimulation. Our findings suggest that SFN improves metabolic changes induced by SE which have been identified during epileptogenesis in various animal models of acquired epilepsy.

3.
Front Neurosci ; 15: 634378, 2021.
Article in English | MEDLINE | ID: mdl-33746702

ABSTRACT

The aim of the present study was to elucidate the effect of resveratrol (natural polyphenol) on seizure activity, production of ROS, brain damage and mitochondrial function in the early phase of status epilepticus (SE), induced in immature 12 day-old rats by substances of a different mechanism of action (Li-pilocarpine, DL-homocysteic acid, 4-amino pyridine, and kainate). Seizure activity, production of superoxide anion, brain damage and mitochondrial function were assessed by EEG recordings, hydroethidium method, FluoroJadeB staining and Complex I activity measurement. A marked decrease of complex I activity associated with the acute phase of SE in immature brain was significantly attenuated by resveratrol, given i.p. in two or three doses (25 mg/kg each), 30 min before, 30 or 30 and 60 min after the induction of SE. Increased O2 .- production was completely normalized, brain damage partially attenuated. Since resveratrol did not influence seizure activity itself (latency, intensity, frequency), the mechanism of protection is likely due to its antioxidative properties. The findings have a clinical relevance, suggesting that clinically available substances with antioxidant properties might provide a high benefit as an add-on therapy during the acute phase of SE, influencing also mechanisms involved in the development of epilepsy.

4.
Front Cell Neurosci ; 12: 266, 2018.
Article in English | MEDLINE | ID: mdl-30210297

ABSTRACT

Status epilepticus (SE), especially in immature animals, is known to produce recurrent spontaneous seizures and behavioral comorbidities later in life. The cause of these adverse long-term outcomes is unknown, but it has been hypothesized that free radicals produced by SE may play a role. We tested this hypothesis by treating immature (P25) rats with the free radical scavenger N-tert-butyl-α-phenylnitrone (PBN) at the time of lithium chloride (LiCl)/pilocarpine (PILO)-induced SE. Later, long-term outcomes were assessed. Cognitive impairment (spatial memory) was tested in the Morris water maze (MWM). Emotional disturbances were assessed by the capture test (aggressiveness) and elevated plus maze's (EPM) test (anxiety). Next, the presence and severity of spontaneous seizures were assessed by continuous video/EEG monitoring for 5 days. Finally, immunochemistry, stereology and morphology were used to assess the effects of PBN on hippocampal neuropathology and neurogenesis. PBN treatment modified the long-term effects of SE in varying ways, some beneficial and some detrimental. Beneficially, PBN protected against severe anatomical damage in the hippocampus and associated spatial memory impairment. Detrimentally, PBN treated animals had more severe seizures later in life. PBN also made animals more aggressive and more anxious. Correlating with these detrimental long-term outcomes, PBN significantly modified post-natal neurogenesis. Treated animals had significantly increased numbers of mature granule cells (GCs) ectopically located in the dentate hilus (DH). These results raise the possibility that abnormal neurogenesis may significantly contribute to the development of post-SE epilepsy and behavioral comorbidities.

5.
Mol Neurobiol ; 55(9): 7512-7522, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29427088

ABSTRACT

The presence of oxidative stress in immature brain has been demonstrated during the acute phase of status epilepticus (SE). The knowledge regarding the long periods of survival after SE is not unequivocal, lacking direct evidence. To examine the presence and time profile of oxidative stress, its functional effect on mitochondria and the influence of an antioxidant treatment in immature rats during epileptogenesis, status epilepticus (SE) was induced in immature 12-day-old rats by Li-pilocarpine and at selected periods of the epileptogenesis; rat pups were subjected to examinations. Hydroethidine method was employed for detection of superoxide anion (O2.-), 3-nitrotyrosine (3-NT), and 4-hydroxynonenal (4-HNE) for oxidative damage of mitochondrial proteins and complex I activity for mitochondrial function. Natural polyphenolic antioxidant resveratrol was given in two schemes: "acute treatment," i.p. administration 30 min before, 30 and 60 min after induction of SE and "full treatment" when applications continued once daily for seven consecutive days (25 mg/kg each dose). The obtained results clearly document that the period of epileptogenesis studied (up to 4 weeks) in immature brain is associated with the significant enhanced production of O2.-, the increased levels of 3-NT and 4-HNE and the persisting deficiency of complex I activity. Application of resveratrol either completely prevented or significantly reduced markers both of oxidative stress and mitochondrial dysfunction. The findings suggest that targeting oxidative stress in combination with current antiepileptic therapies may provide a benefit in the treatment of epilepsy.


Subject(s)
Brain/pathology , Mitochondria/pathology , Oxidative Stress/drug effects , Resveratrol/pharmacology , Status Epilepticus/pathology , Animals , Behavior, Animal/drug effects , Biomarkers/metabolism , Electron Transport Complex I/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Rats, Wistar , Superoxides/metabolism , Survival Analysis
6.
Front Cell Neurosci ; 10: 136, 2016.
Article in English | MEDLINE | ID: mdl-27303267

ABSTRACT

Epilepsy is a neurologic disorder, particularly frequent in infants and children where it can lead to serious consequences later in life. Oxidative stress and mitochondrial dysfunction are implicated in the pathogenesis of many neurological disorders including epilepsy in adults. However, their role in immature epileptic brain is unclear since there have been two contrary opinions: oxidative stress is age-dependent and does not occur in immature brain during status epilepticus (SE) and, on the other hand, evidence of oxidative stress in immature brain during a specific model of SE. To solve this dilemma, we have decided to investigate oxidative stress following SE induced in immature 12-day-old rats by three substances with a different mechanism of action, namely 4-aminopyridine, LiCl-pilocarpine or kainic acid. Fluoro-Jade-B staining revealed mild brain damage especially in hippocampus and thalamus in each of the tested models. Decrease of glucose and glycogen with parallel rises of lactate clearly indicate high rate of glycolysis, which was apparently not sufficient in 4-AP and Li-Pilo status, as evident from the decreases of PCr levels. Hydroethidium method revealed significantly higher levels of superoxide anion (by ∼60%) in the hippocampus, cerebral cortex and thalamus of immature rats during status. SE lead to mitochondrial dysfunction with a specific pronounced decrease of complex I activity that persisted for a long period of survival. Complexes II and IV activities remained in the control range. Antioxidant treatment with SOD mimetic MnTMPYP or peroxynitrite scavenger FeTPPS significantly attenuated oxidative stress and inhibition of complex I activity. These findings bring evidence that oxidative stress and mitochondrial dysfunction are age and model independent, and may thus be considered a general phenomenon. They can have a clinical relevance for a novel approach to the treatment of epilepsy, allowing to target the mechanisms which play a crucial or additive role in the pathogenesis of epilepsies in infants and children.

7.
Int Rev Neurobiol ; 114: 209-43, 2014.
Article in English | MEDLINE | ID: mdl-25078504

ABSTRACT

Epilepsy is one of the most common neurologic disorders affecting a substantial part of the population worldwide. Epileptic seizures represent the situation of increased neuronal activity associated with the enhanced demands for sufficient energy supply. For that purpose, very efficient regulatory mechanisms have to operate to ensure that cerebral blood flow, delivery of oxygen, and nutrients are continuously adapted to the local metabolic needs. The sophisticated regulation has to function in concert at several levels (systemic, tissue, cellular, and subcellular). Particularly, mitochondria play a key role not only in the energy production, but they are also central to many other processes including those leading to neuronal death. Impairment of any of the involved pathways can result in serious functional alterations, neurodegeneration, and potentially in epileptogenesis. The present review will address some of the important issues concerning vascular and metabolic changes in pathophysiology of epilepsy.


Subject(s)
Brain/metabolism , Epilepsy/complications , Metabolic Diseases/etiology , Brain/pathology , DNA, Mitochondrial/genetics , Epilepsy/genetics , Humans , Metabolic Diseases/genetics
8.
Int J Dev Neurosci ; 31(2): 123-30, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23238024

ABSTRACT

We have recently demonstrated the evidence of oxidative stress in brain of immature rats during seizures induced by DL-homocysteic acid (DL-HCA). The aim of the present study was to investigate the antioxidant defense mechanisms under these conditions. Seizures were induced in immature 12-day-old rats by bilateral icv infusion of DL-HCA (600 nmol/side), and the activities of the main antioxidant enzymes were examined in supernatants of the cerebral cortex during the acute phase of seizures and at several periods of survival, up to 5 weeks, following these seizures. In control animals individual antioxidant enzymes revealed different changes during the studied postnatal period (PD 12 till PD 47). Total superoxide dismutase (SOD), CuZn SOD (SOD1), Mn SOD (SOD2) and glutathione peroxidase (GPX) activities were increasing while, catalase activity decreased and the activity of glutathione reductase (GR) remained unchanged. In HCA-treated animals, the activity of total SOD, SOD1 and particularly SOD2 significantly increased at 20 h and 6 days of survival. Importantly, upregulation of SOD2 was also confirmed in mitochondria at the protein level by immunoblotting. The activities of other antioxidant enzymes including catalase and GPX did not significantly differ upon HCA treatment from the appropriate controls at any of the studied time intervals. The pronounced and selective upregulation of SOD2 points to enhanced ROS levels in the mitochondrial matrix. This may be associated with inhibition of respiratory chain complex I that we have demonstrated in our previous studies. The present findings suggest that oxidative stress occurring in the brain of immature rats during and following the seizures induced by DL-HCA is apparently due to both the increased free radical production and the limited antioxidant defense.


Subject(s)
Aging/metabolism , Antioxidants/metabolism , Cerebral Cortex/enzymology , Reactive Oxygen Species/metabolism , Seizures/enzymology , Superoxide Dismutase/metabolism , Animals , Oxidative Stress , Rats , Rats, Wistar , Tissue Distribution , Up-Regulation
9.
Mitochondrion ; 12(1): 35-40, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21530687

ABSTRACT

Mitochondrial dysfunction has been identified as one potential cause of epileptic seizures. Impaired mitochondrial function has been reported for the seizure focus of patients with temporal lobe epilepsy and Ammon's horn sclerosis and of adult and immature animal models of epilepsy. Since mitochondrial oxidative phosphorylation provides the major source of ATP in neurons and mitochondria participate in cellular Ca(2+) homeostasis and generation of reactive oxygen species, their dysfunction strongly affects neuronal excitability and synaptic transmission. Therefore, mitochondrial dysfunction is proposed to be highly relevant for seizure generation. Additionally, mitochondrial dysfunction is known to trigger neuronal cell death, which is a prominent feature of therapy-resistant epilepsy. For this reason mitochondria have to be considered as promising targets for neuroprotective strategies in epilepsy.


Subject(s)
Epilepsy/physiopathology , Mitochondria/physiology , Neurons/physiology , Animals , Calcium/metabolism , Disease Models, Animal , Energy Metabolism , Homeostasis , Humans , Reactive Oxygen Species/metabolism
10.
Exp Neurol ; 233(1): 421-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22108622

ABSTRACT

The widely-held assumption was that oxidative stress does not occur during seizures in the immature brain. The major finding of the present study concerns evidence of oxidative stress in the brain of immature rats during seizures induced by DL-homocysteic acid. Seizures were induced in 12-day-old rats by bilateral intracerebroventricular infusion of DL-homocysteic acid (DL-HCA, 600 nmol/side) and oxidative stress was evaluated by in situ detection of superoxide anion (O(2)·(-)). Using hydroethidine (Het) method, the fluorescent signal of the oxidized products of Het (reflecting O(2)·(-) production) significantly increased (by 50%-60%) following 60 min lasting seizures in all the studied structures, namely CA1, CA3 and dentate gyrus of the hippocampus, cerebral cortex and thalamus. The enhanced O(2)·(-) production was substantially attenuated or completely prevented by substances providing an anticonvulsant effect, namely by a competitive NMDA receptor antagonist AP7, a highly selective and potent group II metabotropic glutamate receptor (mGluR) agonist 2R,4R-APDC and highly selective group III mGluR, subtype 8 agonist (S)-3,4-DCPG. Complete protection was achieved by two SOD mimetics Tempol and MnTMPYP which strongly suggest that the increased fluorescent signal reflects O(2)·(-) formation. In addition, both scavengers provided a partial protection against brain damage associated with the present model of seizures. Signs of neuronal degeneration, as evaluated by Fluoro-Jade B staining, were detected at 4h following the onset of seizures. The present findings thus suggest that the increased superoxide generation precedes neuronal degeneration and may thus play a causative role in neuronal injury. Occurrence of oxidative stress in brain of immature rats during seizures, as demonstrated in the present study, can have a clinical relevance for a novel approach to the treatment of epilepsy in children, suggesting that substances with antioxidant properties combined with the conventional therapies might provide a beneficial effect.


Subject(s)
Anticonvulsants/therapeutic use , Brain/metabolism , Seizures/pathology , Seizures/prevention & control , Superoxides/metabolism , 2-Amino-5-phosphonovalerate/analogs & derivatives , 2-Amino-5-phosphonovalerate/therapeutic use , Animals , Animals, Newborn , Brain/drug effects , Disease Models, Animal , Homocysteine/analogs & derivatives , Homocysteine/toxicity , Infusions, Intraventricular , Male , Metalloporphyrins/metabolism , Proline/analogs & derivatives , Proline/therapeutic use , Rats , Rats, Wistar , Seizures/chemically induced , Statistics, Nonparametric , Time Factors
11.
Neurochem Int ; 56(3): 394-403, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19931336

ABSTRACT

Our previous work demonstrated the marked decrease of mitochondrial complex I activity in the cerebral cortex of immature rats during the acute phase of seizures induced by bilateral intracerebroventricular infusion of dl-homocysteic acid (600 nmol/side) and at short time following these seizures. The present study demonstrates that the marked decrease ( approximately 60%) of mitochondrial complex I activity persists during the long periods of survival, up to 5 weeks, following these seizures, i.e. periods corresponding to the development of spontaneous seizures (epileptogenesis) in this model of seizures. The decrease was selective for complex I and it was not associated with changes in the size of the assembled complex I or with changes in mitochondrial content of complex I. Inhibition of complex I was accompanied by a parallel, up to 5 weeks lasting significant increase (15-30%) of three independent mitochondrial markers of oxidative damage, 3-nitrotyrosine, 4-hydroxynonenal and protein carbonyls. This suggests that oxidative modification may be most likely responsible for the sustained deficiency of complex I activity although potential role of other factors cannot be excluded. Pronounced inhibition of complex I was not accompanied by impaired ATP production, apparently due to excess capacity of complex I documented by energy thresholds. The decrease of complex I activity was substantially reduced by treatment with selected free radical scavengers. It could also be attenuated by pretreatment with (S)-3,4-DCPG (an agonist for subtype 8 of group III metabotropic glutamate receptors) which had also a partial antiepileptogenic effect. It can be assumed that the persisting inhibition of complex I may lead to the enhanced production of reactive oxygen and/or nitrogen species, contributing not only to neuronal injury demonstrated in this model of seizures but also to epileptogenesis.


Subject(s)
Cerebral Cortex/metabolism , Electron Transport Complex I/metabolism , Epilepsy/metabolism , Mitochondrial Diseases/metabolism , Seizures/metabolism , Aldehydes/metabolism , Animals , Animals, Newborn , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Convulsants/toxicity , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/physiology , Electron Transport Complex I/drug effects , Energy Metabolism/drug effects , Energy Metabolism/physiology , Epilepsy/physiopathology , Excitatory Amino Acid Agonists/pharmacology , Free Radical Scavengers/pharmacology , Homocysteine/analogs & derivatives , Homocysteine/toxicity , Male , Metabolic Networks and Pathways/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/chemically induced , Mitochondrial Diseases/physiopathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/physiopathology , Survival Rate , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism
12.
Brain Res ; 1273: 144-54, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19341715

ABSTRACT

The present study has examined the anticonvulsant and neuroprotective effect of 2R,4R-4-aminopyrrolidine-2,4-dicarboxylate (2R,4R-APDC), a selective agonist for group II metabotropic glutamate receptors (mGluRs) when given 10-15 min after the onset of seizures induced in 12-day-old rats by bilateral icv infusion of DL-homocysteic acid (DL-HCA, 600 nmol/side). For biochemical analyses, rat pups were sacrificed during generalized clonic-tonic seizures, approximately 45-50 min after infusion of DL-HCA. Comparable time intervals were used for sacrificing the animals which received 2R,4R-APDC (0.05 nmol/side) or saline. The severity of seizures was influenced only slightly when the agonist was given after the onset of seizures, as evaluated both from the behavioral symptoms and from EEG recordings. A tendency to lower number and a shorter duration of seizures was outlined in animals posttreated with 2R,4R-APDC, but the differences did not reach the level of statistical significance. Cortical energy metabolite changes which normally accompany seizures in immature rats (large decrease of glucose and glycogen and a marked rise of lactate) were ameliorated only partially. The neuroprotective effect of 2R,4R-APDC was evaluated after 24 h and 6 days of survival following DL-HCA-induced seizures. Massive neuronal degeneration in many brain regions, mainly in the hippocampus and thalamus, following infusion of DL-HCA alone was only partially attenuated after 2R,4R-APDC posttreatment. The present findings clearly indicate that both anticonvulsant and neuroprotective effect of 2R,4R-APDC against DL-HCA-induced seizures is substantially diminished when the agonist is given after the onset of seizures as compared with its efficacy after the pretreatment (Exp. Neurol.192, 420-436, 2005).


Subject(s)
Brain/drug effects , Epilepsy/drug therapy , Excitatory Amino Acid Agonists/therapeutic use , Neuroprotective Agents/therapeutic use , Proline/analogs & derivatives , Receptors, Metabotropic Glutamate/agonists , Aging/metabolism , Animals , Brain/growth & development , Brain/metabolism , Convulsants/pharmacology , Cytoprotection/drug effects , Cytoprotection/physiology , Drug Administration Schedule , Drug Interactions/physiology , Epilepsy/metabolism , Epilepsy/physiopathology , Hippocampus/drug effects , Hippocampus/growth & development , Hippocampus/metabolism , Homocysteine/analogs & derivatives , Homocysteine/pharmacology , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Proline/therapeutic use , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/metabolism , Thalamus/drug effects , Thalamus/growth & development , Thalamus/metabolism , Treatment Outcome
13.
Neuropharmacology ; 54(4): 665-75, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18191956

ABSTRACT

The present study has examined the anticonvulsant and neuroprotective effect of (S)-3,4-dicarboxyphenylglycine ((S)-3,4-DCPG), a highly selective agonist for subtype 8 of group III metabotropic glutamate receptors (mGluRs), against seizures induced in immature 12-day-old rats by bilateral icv infusion of DL-homocysteic acid (DL-HCA, 600 nmol/side). For biochemical analyses, rat pups were sacrificed during generalized clonic-tonic seizures, approximately 45-50 min after infusion. Comparable time intervals were used for sacrificing the animals which had received (S)-3,4-DCPG (0.25 nmol/each side, 15-20 min prior to infusion of DL-HCA or saline). This agonist provided a pronounced anticonvulsant effect, generalized clonic-tonic seizures were completely suppressed and cortical energy metabolite changes which normally accompany these seizures were either normalized (decrease of glucose and glycogen) or markedly reduced (an accumulation of lactate). Anticonvulsant effect of (S)-3,4-DCPG was also evident from the EEG recordings, nevertheless, it was not complete. In spite of the absence of obvious motor phenomena, sporadic ictal activity could be seen in some animals. Isolated spikes could also be observed in some animals after administration of (S)-3,4-DCPG alone. The neuroprotective effect of (S)-3,4-DCPG was evaluated after 24 h and 6 days of survival following DL-HCA-induced seizures. Massive neuronal degeneration was observed in a number of brain regions following infusion of DL-HCA alone (seizure group), whereas pretreatment with (S)-3,4-DCPG provided substantial neuroprotection. The present findings suggest that receptor subtype 8 of group III mGluRs may be considered a promising target for drug therapy in childhood epilepsies in the future.


Subject(s)
Anticonvulsants/therapeutic use , Benzoates/therapeutic use , Glycine/analogs & derivatives , Homocysteine/analogs & derivatives , Seizures/chemically induced , Seizures/drug therapy , Animals , Animals, Newborn , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Dose-Response Relationship, Drug , Drug Interactions , Electroencephalography/methods , Fluoresceins , Glycine/therapeutic use , Male , Organic Chemicals , Random Allocation , Rats , Rats, Wistar , Seizures/pathology , Time Factors
14.
Exp Neurol ; 204(2): 597-609, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17270175

ABSTRACT

The major finding of the present study concerns the marked decrease of respiratory chain complex I activity in the cerebral cortex of immature rats following seizures induced by bilateral intracerebroventricular infusion of dl-homocysteic acid (600 nmol/side). This decrease was already evident during the acute phase of seizures (60-90 min after infusion) and persisted for at least 20 h after the seizures. It was selective for complex I since activities of complex II and IV and citrate synthase remained unaffected. Inhibition of complex I activity was not associated with changes in complex I content. Based on enhanced lipoperoxidation and decreased aconitase activity, it can be postulated that oxidative modification is most likely responsible for the observed inhibition. Mitochondrial respiration, as well as cortical ATP levels remained in the control range, apparently due to excess capacity of the complex I documented by energy thresholds. On the other hand, the enhanced production of reactive oxygen species by inhibited complex I was observed in mitochondria from HCA-treated animals. The decrease of complex I activity was substantially attenuated when animals were treated with substances providing an anticonvulsant effect and also with selected free radical scavengers. We can assume that inhibition of complex I may elicit enhanced formation of reactive oxygen species and contribute thus to neuronal injury demonstrated in this model.


Subject(s)
Cerebral Cortex/enzymology , Electron Transport Complex I/metabolism , Homocysteine/analogs & derivatives , Seizures/pathology , Aconitate Hydratase/metabolism , Analysis of Variance , Animals , Animals, Newborn , Antioxidants/pharmacology , Cerebral Cortex/growth & development , Cerebral Cortex/ultrastructure , Citrate (si)-Synthase/metabolism , Cyclic N-Oxides/pharmacology , Drug Interactions , Electron Transport Complex I/antagonists & inhibitors , Energy Metabolism/drug effects , Lipid Peroxidation/drug effects , Male , Metalloporphyrins/pharmacology , Mitochondria/drug effects , Oxygen Consumption/drug effects , Rats , Rats, Wistar , Seizures/chemically induced , Spin Labels
15.
Exp Neurol ; 201(1): 105-19, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16756975

ABSTRACT

The present study has examined the effect of free radical spin trap N-tert-butyl-alpha-phenylnitrone (PBN) in the model of seizures induced in immature 12-day-old rats by bilateral intracerebroventricular infusion of dl-homocysteic acid (dl-HCA, 600 nmol/side). PBN was given i.p. in two doses (100 mg/kg each), 30 min prior and 30 min after dl-HCA infusion. PBN did not significantly influence the severity of seizures, evident both from the behavioral symptoms and EEG recordings. PBN normalized decreased ATP levels in the hippocampus, occurring during the acute phase of seizures ( approximately 45-50 min after infusion) and persisting until the end of the 24-h recovery period. PBN also led to normalization of decreased glucose levels and to a significant reduction of lactate accumulation in the cerebral cortex and hippocampus. The neuroprotective effect of PBN was evaluated after 24 h and 6 days of survival following dl-HCA-induced seizures (Nissl and Fluoro-Jade B staining). The administration of PBN resulted in a partial amelioration of severe damage observed in many brain regions following infusion of dl-HCA alone. The data suggest that increased free radical production is apparently occurring during seizures induced in immature rats by homocysteic acid. Free radical scavenger PBN had a clear-cut protective effect, evident as the improved recovery of brain energy status and as a partial, but significant, attenuation of neuronal degeneration associated with this model of seizures.


Subject(s)
Brain/drug effects , Disaccharides/pharmacology , Seizures/prevention & control , Adenosine Triphosphate/metabolism , Animals , Brain/metabolism , Brain/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disaccharides/administration & dosage , Electroencephalography , Energy Metabolism/drug effects , Free Radicals/antagonists & inhibitors , Glucose/metabolism , Glycogen/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Homocysteine/analogs & derivatives , Homocysteine/toxicity , Injections, Intraperitoneal , Male , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Phosphocreatine/metabolism , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/physiopathology , Spin Trapping
16.
Exp Neurol ; 192(2): 420-36, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15755559

ABSTRACT

The present study has examined the anticonvulsant and neuroprotective effect of group II metabotropic glutamate receptor (mGluR) agonist (2R,4R)-4-aminopyrrolidine-2,4-dicarboxylate (2R,4R-APDC) in the model of seizures induced in immature 12-day-old rats by bilateral intracerebroventricular infusion of dl-homocysteic acid (DL-HCA, 600 nmol/side). For biochemical analyses, rat pups were sacrificed during generalized clonic-tonic seizures, approximately 45-50 min after infusion. Comparable time intervals were used for sacrificing the pups which had received 2R,4R-APDC. Low doses of 2R,4R-APDC (0.05 nmol/side) provided a pronounced anticonvulsant effect which was abolished by pretreatment with a selective group II mGluR antagonist LY341495. Generalized clonic-tonic seizures were completely suppressed and cortical energy metabolite changes which normally accompany these seizures were either normalized (decrease of glucose and glycogen) or markedly reduced (an accumulation of lactate). EEG recordings support the marked anticonvulsant effect of 2R,4R-APDC, nevertheless, this was only partial. In spite of the absence of obvious motor phenomena, isolated spikes or even short periods of partial ictal activity could be observed. Isolated spikes could also be seen in some animals after application of 2R,4R-APDC alone, reflecting most likely subclinical proconvulsant activity of this agonist. The neuroprotective effect of 2R,4R-APDC was evaluated after 24 h and 6 days of survival following DL-HCA-induced seizures. Massive neuronal degeneration, as revealed by Fluoro-Jade B staining, was observed in a number of brain regions following infusion of DL-HCA alone (seizure group), whereas 2R,4R-APDC pretreatment provided substantial neuroprotection. The present findings support the possibility that group II mGluRs are a promising target for a novel approach to treating epilepsy.


Subject(s)
Brain Injuries/etiology , Brain Injuries/prevention & control , Homocysteine/analogs & derivatives , Proline/analogs & derivatives , Proline/therapeutic use , Receptors, Metabotropic Glutamate/agonists , Seizures/complications , Amino Acids/pharmacology , Animals , Animals, Newborn , Anticonvulsants/therapeutic use , Behavior, Animal , Brain/anatomy & histology , Brain/drug effects , Brain/physiopathology , Brain Chemistry/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Electroencephalography/methods , Excitatory Amino Acid Antagonists/pharmacology , Fluoresceins , Fluorescent Dyes , Functional Laterality , Glucose/metabolism , Glycogen/metabolism , Lactic Acid/metabolism , Male , Nerve Degeneration/pathology , Nerve Degeneration/prevention & control , Organic Chemicals , Rats , Rats, Wistar , Seizures/chemically induced , Time Factors , Xanthenes/pharmacology
17.
Exp Neurol ; 180(1): 46-54, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12668148

ABSTRACT

The potential anticonvulsant effect of group III metabotropic glutamate receptor (mGluR) agonist (R,S)-4-phosphonophenylglycine ((R,S)-PPG) against seizures induced in immature 12-day-old rats by bilateral intracerebroventricular (icv) infusion of DL-homocysteic acid (DL-HCA, 600 nmol/side) was examined in the present study. Rat pups were sacrificed during generalized clonic-tonic seizures, approximately 45 to 50 min after infusion. Comparable time intervals were used for sacrificing the pups which had received (R,S)-PPG. Low doses of (R,S)-PPG (10 nmol, icv) provided a pronounced anticonvulsant effect which was abolished by pretreatment with a selective group III mGluR antagonist (R,S)-alpha-methylserine-O-phosphate. Generalized clonic-tonic seizures were completely suppressed and cortical energy metabolite changes which normally accompany these seizures were either normalized (glucose and glycogen decreases) or markedly ameliorated (an accumulation of lactate). Despite the absence of obvious motor phenomena, EEG recordings revealed sporadic ictal activity, mostly in the dorsal hippocampus. Spreading of this activity into the frontal cortex was rather exceptional. The latency of ictal EEG in pretreated rats was significantly prolonged. Our data suggest that the predominant effect of (R,S)-PPG might concern seizure spread. The administration of (R,S)-PPG alone did not cause any overt behavioral side effects; it did not change the EEG pattern and did not influence cortical metabolite levels, with the exception of increased concentrations of glucose. The present findings suggest that group III mGlu receptor agonists may be of therapeutic significance for treating childhood epilepsies.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Homocysteine/analogs & derivatives , Receptors, Metabotropic Glutamate/agonists , Seizures/chemically induced , Seizures/prevention & control , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Electroencephalography , Excitatory Amino Acid Antagonists/pharmacology , Frontal Lobe/drug effects , Frontal Lobe/physiopathology , Glucose/metabolism , Hippocampus/drug effects , Hippocampus/physiopathology , Injections, Intraventricular , Male , Phosphoserine/pharmacology , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Seizures/physiopathology
18.
Epilepsia ; 44(3): 299-304, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12614384

ABSTRACT

PURPOSE: To examine the morphologic alterations in the cerebral cortex and hippocampus of immature rats 6 days after the generalized clonic-tonic seizures induced by homocysteic acid (HCA). METHODS: Seizures were induced by bilateral intracerebroventricular infusion of HCA (600 nmol per each side) in 12-day-old rats. After 6 days, rat pups were transcardially perfused under deep ether anesthesia with heparinized normal saline and subsequently with the fixation solution (4% paraformaldehyde in phosphate buffer, pH 7.4, for light microscopy) or with Karnovsky's solution (4% paraformaldehyde and 2% glutaraldehyde in phosphate buffer, pH 7.4, for electron microscopic analysis). Nissl stain and the DNA-specific dye bis-benzimide (Hoechst 33342) were used. RESULTS: No pathologic changes were found in the cerebral cortex, whereas serious alterations occurred in the hippocampus. A total loss of CA3 pyramidal cells was observed, with marked changes in the CA1 region and dentate gyrus. A prominent glial reaction was seen in many regions of the hippocampal formation. A slight dilatation of the cerebral ventricles was noticed in some experimental as well as control animals. In the granule cell layer of the dentate gyrus, neurons with segmented or fragmented nuclei in various stages of degeneration were detected, displaying the features of apoptotic death. CONCLUSIONS: These findings demonstrate the vulnerability of the immature rat brain, which most likely reflects both the direct neurotoxic effect of HCA and prolonged seizure activity. The relative contribution of these two factors still remains to be assessed.


Subject(s)
Apoptosis , Hippocampus/drug effects , Hippocampus/pathology , Homocysteine , Homocysteine/analogs & derivatives , Neurons/pathology , Seizures/chemically induced , Animals , Animals, Newborn , Apoptosis/physiology , Cell Count , Cell Nucleus/pathology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Hippocampus/cytology , Homocysteine/pharmacology , Male , Nerve Degeneration/pathology , Neuroglia/cytology , Neuroglia/pathology , Neurons/cytology , Neurons/drug effects , Pyramidal Cells/cytology , Pyramidal Cells/pathology , Rats , Rats, Wistar , Seizures/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...