Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Epilepsia ; 49(10): 1759-76, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18494789

ABSTRACT

PURPOSE: Patients with intractable temporal lobe epilepsy (TLE) exhibit an increased risk of psychiatric comorbidity, including depression, anxiety, psychosis, and learning disorders. Furthermore, a history of psychiatric comorbidity has been suggested as a predictor of lack of response to therapy with antiepileptic drugs (AEDs) in patients with epilepsy. However, clinical studies on predictors of pharmacoresistant epilepsy are affected by several confounding variables, which may complicate conclusions. In the present study, we evaluated whether behavioral alterations in epileptic rats are different in AED nonresponders versus responders. METHODS: For this purpose, we used an animal model of TLE in which AED responders and nonresponders can be selected by prolonged treatment of epileptic rats with phenobarbital (PB). Behavioral and cognitive abnormalities were compared between responders and nonresponders as well as between epileptic rats and nonepileptic controls in a battery of tests. RESULTS: Fifteen epileptic rats with spontaneous recurrent seizures (SRS) either responding (11 rats) or not responding (4 rats) to PB were used for this study. The nonresponders differed markedly in behavioral and cognitive abnormalities from responders and nonepileptic controls in tests of anxiety (open field, elevated-plus maze test), behavioral hyperexcitability (approach-response, touch-response, pick-up tests), and learning and memory (Morris water maze). DISCUSSION: Our hypothesis that AED-resistant rats will show more severe behavioral and cognitive changes than AED-responsive rats was confirmed by the present experiments. The data substantiate that rodent models of TLE are useful to delineate predictors of pharmacoresistant epilepsy.


Subject(s)
Anticonvulsants/therapeutic use , Behavior, Animal/drug effects , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Drug Resistance , Epilepsy/complications , Amygdala/physiology , Amygdala/radiation effects , Analysis of Variance , Animals , Disease Models, Animal , Drug Resistance/drug effects , Electric Stimulation/adverse effects , Electroencephalography/methods , Epilepsy/etiology , Epilepsy/pathology , Epilepsy/prevention & control , Exploratory Behavior/drug effects , Female , Hippocampus/pathology , Maze Learning/drug effects , Phenobarbital/therapeutic use , Predictive Value of Tests , Rats , Rats, Sprague-Dawley , Secondary Prevention , Statistics, Nonparametric , Swimming/physiology , Time Factors
2.
Neuropharmacology ; 53(2): 207-21, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17585956

ABSTRACT

Levetiracetam (LEV) is a structurally novel antiepileptic drug (AED) which has demonstrated a broad spectrum of anticonvulsant activities both in experimental and clinical studies. Previous experiments in the kindling model suggested that LEV, in addition to its seizure-suppressing activity, may possess antiepileptogenic or disease-modifying activity. In the present study, we evaluated this possibility by using a rat model in which epilepsy with spontaneous recurrent seizures (SRS), behavioral alterations, and hippocampal damages develop after a status epilepticus (SE) induced by sustained electrical stimulation of the basal amygdala. Two experimental protocols were used. In the first protocol, LEV treatment was started 24h after onset of electrical amygdala stimulation without prior termination of the SE. In the second protocol, the SE was interrupted after 4h by diazepam, immediately followed by onset of treatment with LEV. Treatment with LEV was continued for 8 weeks (experiment #1) or 5 weeks (experiment #2) after SE, using continuous drug administration via osmotic minipumps. The occurrence of SRS was recorded during and after treatment. In addition, the rats were tested in a battery of behavioral tests, including the elevated-plus maze and the Morris water maze. Finally, the brains of the animals were analyzed for histological lesions in the hippocampal formation. With the experimental protocols chosen for these experiments, LEV did not exert antiepileptogenic or neuroprotective activity. Furthermore, the behavioral alterations, e.g., behavioral hyperexcitability and learning deficits, in epileptic rats were not affected by treatment with LEV after SE. These data do not support the idea that administration of LEV after SE prevents or reduces the long-term alterations developing after such brain insult in rats.


Subject(s)
Anticonvulsants/administration & dosage , Behavior, Animal/drug effects , Brain Damage, Chronic/prevention & control , Piracetam/analogs & derivatives , Status Epilepticus/drug therapy , Amygdala/physiology , Amygdala/radiation effects , Analysis of Variance , Animals , Brain Damage, Chronic/pathology , Diazepam/administration & dosage , Disease Models, Animal , Drug Administration Schedule , Electric Stimulation/adverse effects , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , Hippocampus/drug effects , Hippocampus/pathology , Hyperkinesis/drug therapy , Hyperkinesis/etiology , Levetiracetam , Maze Learning/drug effects , Maze Learning/physiology , Piracetam/administration & dosage , Rats , Rats, Sprague-Dawley , Status Epilepticus/complications , Status Epilepticus/etiology , Status Epilepticus/pathology , Swimming/psychology , Time Factors
3.
Neuropharmacology ; 52(2): 333-46, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17045309

ABSTRACT

In view of the important role of P-glycoprotein (Pgp) and other drug efflux transporters for drug distribution and resistance, the identification of compounds as substrates of Pgp-mediated transport is one of the key issues in drug discovery and development, particularly for compounds acting on the central nervous system. In vitro transport assays with Pgp-transfected kidney cell lines are widely used to evaluate the potential of compounds to act as Pgp substrates or inhibitors. Furthermore, such cell lines are also frequently utilized as a substitute for more labor-intensive in vitro or in vivo models of the blood-brain barrier (BBB). Overexpression of Pgp or members of the multidrug resistance protein (MRP) family at the BBB has been implicated in the mechanisms underlying resistance to antiepileptic drugs (AEDs) in patients with epilepsy. Therefore, it is important to know which AEDs are substrates for Pgp or MRPs. In the present study, we used monolayers of polarized MDCKII dog kidney or LLC-PK1 pig kidney cells transfected with cDNA containing either human MDR1, MRP2 or mouse mdr1a and mdr1b sequences to measure the directional transport of AEDs. Cyclosporin A (CsA) and vinblastine were used as reference standards for Pgp and MRP2, respectively. The AEDs phenytoin and levetiracetam were directionally transported by mouse but not human Pgp, whereas CsA was transported by both types of Pgp. Carbamazepine was not transported by any type of Pgp and did not inhibit the transport of CsA. In contrast to vinblastine, none of the AEDs was transported by MRP2 in transfected kidney cells. The data indicate that substrate recognition or transport efficacy by Pgp differs between human and mouse for certain AEDs. Such species differences, which are certainly not restricted to human and mouse, may explain, at least in part, the controversial data which have been previously reported for AED transport by Pgp in preparations from different species. However, because transport efficacy of efflux transporters such as Pgp or MRP2 may not only differ between species but also between tissues, the present data do not exclude that the AEDs examined are weak substrates of Pgp or MRP2 at the human BBB.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Anticonvulsants/pharmacokinetics , Carbamazepine/pharmacokinetics , Phenytoin/pharmacokinetics , Piracetam/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Analysis of Variance , Animals , Biological Transport/drug effects , Cell Line , Cyclosporine/pharmacology , Dogs , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Humans , Levetiracetam , Mice , Piracetam/pharmacokinetics , Transfection
4.
Neurobiol Dis ; 24(1): 202-11, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16928449

ABSTRACT

Resistance to drug treatment is an important hurdle in the therapy of many diseases, including cancer, infectious diseases and brain disorders such as epilepsy. A phenotype that is referred to as multidrug resistance was first described for chemotherapy-resistant cancer cells that overexpressed the drug efflux transporter P-glycoprotein (P-gp). More recently, overexpression of P-gp has been found in capillary endothelial cells of epileptogenic brain tissue from patients with medically intractable epilepsy. Such regionally restricted P-gp overexpression in the blood-brain barrier is likely to reduce the concentration of antiepileptic drugs at epileptic neurons, which would be a plausible explanation for multidrug resistance in epilepsy. However, a definite proof-of-principle for this hypothesis is lacking. In the present study, we used a rat model of temporal lobe epilepsy that allows selecting drug-resistant and drug-responsive subgroups of epileptic rats by prolonged treatment with the antiepileptic drug phenobarbital at maximum tolerated doses. We have shown recently that drug-resistant rats selected from this model exhibit a marked overexpression of P-gp in the hippocampus and other limbic brain regions. This model is thus ideally suited to prove the multidrug transporter hypothesis of drug resistance. For this purpose, we selected a group of phenobarbital-resistant rats, which was subsequently treated by combinations of phenobarbital with the selective P-gp inhibitor tariquidar. Coadministration of tariquidar (15-20 mg/kg) fully restored the anticonvulsant activity of phenobarbital without altering plasma pharmacokinetics or neurotoxicity of the antiepileptic drug. These data demonstrate that inhibiting P-gp in epileptic rats with proven drug resistance counteracts resistance, providing the first proof-of-principle of the multidrug transporter hypothesis of medically refractory epilepsy.


Subject(s)
Anticonvulsants/pharmacology , Carrier Proteins/genetics , Drug Resistance, Multiple/genetics , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Anticonvulsants/pharmacokinetics , Electrodes, Implanted , Electroencephalography/drug effects , Female , Phenobarbital/pharmacokinetics , Phenobarbital/pharmacology , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Seizures/physiopathology , Stereotaxic Techniques
5.
Neuropharmacology ; 51(4): 789-804, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16806297

ABSTRACT

Epileptogenesis, i.e. the process leading to epilepsy with spontaneous recurrent seizures, can be initiated by a number of brain damaging insults, including traumatic brain injury, status epilepticus (SE), and stroke. Such acquired epilepsy is often associated with memory impairment and behavioral problems. There has been a growing interest in the use of antiepileptic drugs (AEDs) for neuroprotection and prevention or modification of epileptogenesis induced by such brain insults. One promising candidate in this respect is valproic acid (VPA), a widely used AED that has been reported to exert neuroprotective activity in a number of in vitro and in vivo models. The present study investigated whether VPA reduces brain damage and improves functional outcome in a rat model of post-SE epilepsy. A self-sustaining SE was induced by prolonged electrical stimulation of the basal amygdala via a depth electrode. SE was terminated after 4 h by diazepam, immediately followed by onset of treatment with VPA. VPA was injected i.p. at a bolus dose of 400 mg/kg, followed by three times daily administration of 200 mg/kg for 4 weeks. A control group received vehicle instead of VPA after SE. Spontaneous seizures were recorded in all rats of both groups following termination of treatment, without significant inter-group difference in seizure frequency or severity. However, treatment with VPA after SE prevented the hyperexcitability and locomotor hyperactivity observed in vehicle-treated epileptic rats. Furthermore, VPA completely counteracted the neuronal damage in the hippocampal formation, including the dentate hilus. The data demonstrate that, although VPA does not prevent the occurrence of spontaneous seizures after SE, it exerts powerful neuroprotective effects and prevents part of the behavioral alterations, demonstrating that administration of VPA immediately after SE exerts a favorable effect on long-term functional outcome.


Subject(s)
Anticonvulsants/therapeutic use , Behavior, Animal/drug effects , Nerve Degeneration/drug therapy , Status Epilepticus , Valproic Acid/therapeutic use , Animals , Body Weight/drug effects , Cell Death/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation/adverse effects , Electroencephalography/methods , Female , Functional Laterality , Hippocampus/drug effects , Hippocampus/pathology , Hyperkinesis/drug therapy , Hyperkinesis/etiology , Maze Learning/drug effects , Motor Activity/drug effects , Nerve Degeneration/etiology , Rats , Rats, Sprague-Dawley , Rotarod Performance Test/methods , Status Epilepticus/drug therapy , Status Epilepticus/pathology , Status Epilepticus/physiopathology , Time Factors
6.
Epilepsy Res ; 69(1): 1-14, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16504477

ABSTRACT

Multidrug resistance proteins (MRPs; symbol ABCC) are membrane glycoproteins that mediate the ATP-dependent export of a wide range of substrates from cells and thereby affect the bioavailability and disposition of many drugs. MRP2 (ABCC2) is expressed on the apical domain of hepatocytes, enterocytes of the proximal small intestine, and proximal renal tubular cells, but its location in the brain is a matter of debate. Most previous studies failed to determine MRP2 mRNA or protein in the brain or cell preparations from the brain of different species including humans. Based on our previous experience with the drug efflux transporter P-glycoprotein, we evaluated whether the immunohistochemical determination of MRP2 expression is sensitive to fixation and staining variables. Furthermore, we examined whether the MRP2 protein is overexpressed after experimentally induced seizures in rats, using the pilocarpine model of temporal lobe epilepsy. The MRP2 expression in the liver was used as positive control. MRP2 deficient TR- rats were used as negative controls. Despite various modifications in tissue fixation and immunohistochemical staining as well as use of different commercially available MRP2 antibodies, we never observed any unequivocal MRP2 staining in the brain of normal rats. However, after a pilocarpine-induced convulsive status epilepticus, clear MRP2 staining became visible in brain capillary endothelial cells and, less frequently, perivascular astroglia and neurons in various brain regions. In view of our recent data on brain access of antiepileptic drugs in MRP2 deficient TR- rats, seizure-induced over-expression of MRP2 in the blood-brain barrier is likely to impair drug penetration into the brain, thereby contributing to drug resistance in epilepsy.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Epilepsy, Temporal Lobe/metabolism , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Seizures/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antibodies, Monoclonal , Drug Resistance, Multiple , Epilepsy, Temporal Lobe/chemically induced , Female , Immunohistochemistry , Male , Mice , Multidrug Resistance-Associated Protein 2 , Pilocarpine , RNA, Messenger , Rats , Rats, Inbred Strains , Rats, Wistar , Seizures/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...