Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Epilepsy Res ; 108(4): 634-43, 2014 May.
Article in English | MEDLINE | ID: mdl-24656981

ABSTRACT

The glycine co-agonist binding site of the N-methyl-D-aspartat (NMDA) receptor is discussed as an interesting target for different central nervous system diseases. Antagonism at this co-agonist site has been suggested as an alternative to the use of non-competitive or competitive NMDA receptor antagonists, which are associated with a pronounced adverse effect profile in chronic epilepsy models and epilepsy patients. In the present study, we addressed the hypothesis that sub-chronic administration of the glycine-binding site antagonist L-701,324 might exert disease-modifying effects in fully kindled mice during a period with frequent seizure elicitation (massive kindling). Moreover, we analyzed whether L-701,324 exposure during this phase affects the subsequent response to an antiepileptic drug. L-701,324 treatment during the massive kindling phase did not affect ictogenesis. Mean seizure severity and cumulative seizure duration proved to be comparable between vehicle- and L-701,324-treated mice. Following withdrawal of L-701,324 seizure thresholds did not differ in a significant manner from those in animals that received vehicle injections. A low dosage of phenobarbital caused a significant increase of the generalized seizure threshold in the L-701,324 pre-treated group, whereas it did not exert a comparable effect in animals that received vehicle during the massive kindling phase. Analysis of P-glycoprotein in the hilus of the hippocampus revealed lower expression rates in L-701,324 pre-treated kindled mice. In conclusion, the data indicate that targeting of the NMDA receptor glycine-binding site does not result in anticonvulsant or disease-modifying effects. However, it might improve antiepileptic drug responses. The findings might be linked to an impact on P-glycoprotein expression. However, future studies are necessary to further evaluate the mechanisms and assess the potential of respective add-on approaches.


Subject(s)
Anticonvulsants/therapeutic use , Excitatory Amino Acid Antagonists/pharmacology , Kindling, Neurologic/drug effects , Phenobarbital/therapeutic use , Quinolones/pharmacology , Seizures/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Drug Interactions , Male , Mice , Seizures/metabolism
2.
Epilepsy Res ; 108(2): 212-22, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24345645

ABSTRACT

OBJECTIVE: The present study was aimed at determining the effect of eslicarbazepine acetate (ESL), eslicarbazepine and (R)-licarbazepine administration in the mouse corneal kindling and amygdala kindling models. METHODS: NMRI mice were kindled by bilateral corneal stimulation twice daily. In amygdala kindling, mice were stimulated once daily via an implanted depth electrode until 10 generalized seizures were elicited. Maximal electroshocks (MES) were administered via corneal electrodes. RESULTS: The average number of stimulations to reach a fully kindled generalized seizure was markedly increased by ESL. Administration of eslicarbazepine also inhibited the acquisition of kindling, whereas administration of R-licarbazepine did not affect the number of stimulations necessary to induce a specific seizure stage, and did not exert any relevant effect on mean seizure severity during kindling progression. ESL dose-dependently increased the focal seizure threshold and reduced seizure severity in amygdala kindling. Whereas eslicarbazepine treatment increased the afterdischarge threshold in a significant manner, (R)-licarbazepine treatment failed to exert a significant effect on thresholds in fully kindled mice. Administration of ESL and of eslicarbazepine significantly protected mice against MES-induced seizures, whereas that of (R)-licarbazepine failed to provide protection. CONCLUSIONS: These data provide evidence of the anticonvulsant effect of ESL and its active metabolite eslicarbazepine on partial-onset seizures in corneal and amygdala kindling models. Based on an effect of the parent compound and the active metabolite eslicarbazepine, ESL treatment may not merely suppress seizure activity but may also provide a disease-modifying or antiepileptogenic effect. Future studies will be necessary to further evaluate a putative preventive effect, in particular when considering that re-stimulation following wash-out did not indicate a persistent effect. The findings reported here raise doubts on the contribution of (R)-licarbazepine as an active anticonvulsant.


Subject(s)
Amygdala/drug effects , Anticonvulsants/therapeutic use , Cornea/drug effects , Dibenzazepines/therapeutic use , Epilepsy, Temporal Lobe/drug therapy , Kindling, Neurologic/drug effects , Amygdala/physiology , Animals , Anticonvulsants/pharmacology , Cornea/physiology , Dibenzazepines/pharmacology , Dose-Response Relationship, Drug , Epilepsy, Temporal Lobe/physiopathology , Female , Kindling, Neurologic/physiology , Male , Mice
3.
Neurobiol Dis ; 48(3): 454-63, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22750527

ABSTRACT

In the epileptic brain, seizures can increase hippocampal neurogenesis, while opposingly seizure-associated brain pathology has been shown to detrimentally affect neurogenesis. The long-term impact of recurrent seizures on the number of new neurons as well as their relative contribution to the granule cell layer remains an open question. Therefore we analyzed neuron addition based on genetic fate mapping in a chronic model of epilepsy comparing non-kindled animals and kindled animals having at least one generalized seizure with and without further seizures. The number of all new granule cells added to the dentate gyrus following the onset of kindling was significantly increased (7.0-8.9 fold) in kindled groups. The hyperexcitable kindled state and a prior seizure history proved to be sufficient to cause a pronounced long-term net effect on neuron addition. An ongoing continuous occurrence of seizures did not further increase the number of new granule cells in the long-term. In contrast, a correlation was found between the cumulative duration of seizures and neuron addition following a kindled state. In addition, the overall number of seizures influenced the relative portion of new cells among all granule cells. Non-kindled animals showed 1.6% of new granule cells among all granular cells by the end of the experiment. This portion reached 5.7% in the animals which experienced either 10 or 22 seizures. A percentage of 8.4% new cells were determined in the group receiving 46 seizures which is a significant increase in comparison to the control group. In conclusion, permanent genetic fate mapping analysis demonstrated that recurrent seizures result in a lasting change in the makeup of the granule cell layer with alterations in the relative contribution of newborn neurons to the granule cell network. Interestingly, the formation of a hyperexcitable kindled network even without recent seizure activity can result in pronounced long-term alterations in the absolute number of new granule cells. However, seizure density also seems to play a critical role with more frequent seizures resulting in increased fractions of new neurons.


Subject(s)
Dentate Gyrus/pathology , Epilepsy, Temporal Lobe/pathology , Neurogenesis/physiology , Adult Stem Cells/pathology , Animals , Disease Models, Animal , Epilepsy, Temporal Lobe/genetics , Kindling, Neurologic , Mice , Neural Stem Cells/pathology , Neurons , Reverse Transcriptase Polymerase Chain Reaction
4.
Epilepsia ; 52(7): e62-5, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21627644

ABSTRACT

The endocannabinoid system can be considered as a putative target to affect ictogenesis as well as the generation of a hyperexcitable epileptic network. Therefore, we evaluated the effect of a CB1 receptor agonist (WIN55.212-2) and of an inhibitor of the enzymatic degradation of the endocannabinoid anandamide (fatty acid hydrolase inhibitor URB597) in the amygdala kindling model of temporal lobe epilepsy. Only minor effects on seizure thresholds and seizure parameters without a clear dose-dependency were observed in fully kindled mice. When evaluating the impact on kindling acquisition, WIN55.212-2 significantly delayed the progression of seizure severity. In contrast, URB597 did not affect the development of seizures in the kindling paradigm. Analysis of cell proliferation and neurogenesis during the kindling process revealed that URB597 significantly reduced the number of newborn neurons. These data give first evidence that CB1-receptor activation might render a disease-modifying approach. Future studies are necessary that further analyze the role of CB1 receptors and to confirm the efficacy of CB1-receptor agonists in other models of chronic epilepsy.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Amygdala/physiopathology , Benzamides/pharmacology , Benzoxazines/pharmacology , Carbamates/pharmacology , Epilepsy, Temporal Lobe/etiology , Kindling, Neurologic/physiology , Models, Neurological , Morpholines/pharmacology , Naphthalenes/pharmacology , Receptor, Cannabinoid, CB1/agonists , Amygdala/drug effects , Animals , Disease Models, Animal , Epilepsy, Temporal Lobe/physiopathology , Kindling, Neurologic/drug effects , Male , Mice , Seizures/chemically induced , Seizures/physiopathology
5.
Epilepsy Res ; 91(1): 57-65, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20655707

ABSTRACT

The prostaglandin E2 EP1 receptor as well as the inflammatory enzyme cyclooxygenase-2 have been suggested as targets for disease modulation, improvement of therapeutic response, and restoration of pharmacosensitivity in epilepsies. Translational development of respective add-on approaches requires careful analysis of putative effects on ictogenesis. Therefore we evaluated the impact of the EP1 receptor antagonist SC-51089, the EP1 receptor agonist misoprostol and the COX-2 inhibitors celecoxib and NS-398 in the mouse amygdala kindling model of temporal lobe epilepsy. Neither celecoxib nor NS-398 affected the generation, spread and termination of seizure activity. Whereas SC-51089 did not affect the seizure threshold, the highest dose (30mg/kg) significantly decreased the seizure severity when administered 60min before stimulation. Moreover, SC-51089 significantly prolonged seizure duration at the highest dose. The EP1 receptor agonist misoprostol exerted contrasting effects on seizure duration with a significant decrease in the duration of motor seizure activity. The data suggest that doses of COX-2 inhibitors and EP1 receptor antagonists which exert disease modulating or antiepileptic drug potentiating effects do not negatively affect seizure control in temporal lobe epilepsy. The contrasting impact of the EP1 receptor antagonist and agonist suggests that EP1 receptors can influence endogenous mechanisms involved in termination of seizure activity.


Subject(s)
Amygdala/drug effects , Cyclooxygenase 2 Inhibitors/administration & dosage , Cyclooxygenase 2/metabolism , Disease Models, Animal , Drug Delivery Systems , Kindling, Neurologic/drug effects , Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors , Amygdala/metabolism , Animals , Cyclooxygenase 2/physiology , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Hydrazines/administration & dosage , Kindling, Neurologic/physiology , Male , Mice , Oxazepines/administration & dosage , Receptors, Prostaglandin E, EP1 Subtype/physiology
6.
Epilepsia ; 51(9): 1780-90, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20633036

ABSTRACT

PURPOSE: Based on experimental findings, overexpression of P-glycoprotein at the blood-brain barrier has been suggested to be a contributor to pharmacoresistance of the epileptic brain. We test a technique for evaluation of interindividual differences of elevated transporter function, through microPET analysis of the impact of the P-glycoprotein modulator tariquidar. The preclinical study is intended for eventual translation to clinical research of patients with pharmacoresistant seizure disorders. METHODS: We made a microPET evaluation of the effects of tariquidar on the brain kinetics of the P-glycoprotein substrate [(18) F]MPPF in a rat model with spontaneous recurrent seizures, in which it has previously been demonstrated that phenobarbital nonresponders exhibit higher P-glycoprotein expression than do phenobarbital responders. RESULTS: Mean baseline parametric maps of the [(18) F]MPPF unidirectional blood-brain clearance (K(1) ; ml/g per min) and the efflux rate constant (k(2) ; per min) did not differ between the nonresponder and responder group. Tariquidar pretreatment increased the magnitude of [(18) F]MPPF K(1) in hippocampus by a mean of 142% in the nonresponders, which significantly exceeded the 92% increase observed in the responder group. The same treatment decreased the mean magnitude of [(18) F]MPPF k(2) in hippocampus by 27% in nonresponders, without comparable effects in the responder group. DISCUSSION: These results constitute a proof-of-concept for a novel imaging approach to evaluate blood-brain barrier P-glycoprotein function in animals. By extension, [(18) F]MPPF positron emission tomography (PET) with tariquidar pretreatment may be amenable for clinical applications exploring further the relevance of P-glycoprotein overexpression, and for enabling the rational design of pharmacotherapy according to individual differences in P-glycoprotein expression.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Drug Resistance, Multiple/physiology , Epilepsy, Temporal Lobe/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Animals , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/metabolism , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes , Disease Models, Animal , Epilepsy, Temporal Lobe/diagnostic imaging , Epilepsy, Temporal Lobe/drug therapy , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Humans , Phenobarbital/metabolism , Phenobarbital/pharmacology , Phenobarbital/therapeutic use , Positron-Emission Tomography , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Seizures/diagnostic imaging , Seizures/metabolism
7.
Epilepsy Res ; 87(2-3): 177-83, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19767176

ABSTRACT

Pyrrolidine dithiocarbamate (PDTC) has a dual mechanism of action as an antioxidant and an inhibitor of the transcription factor kappa-beta. Both, production of reactive oxygen species as well as activation of NF-kappaB have been implicated in severe neuronal damage in different sub-regions of the hippocampus as well as in the surrounding cortices. The effect of PDTC on status epilepticus-associated cell loss in the hippocampus and piriform cortex was evaluated in the rat fractionated pilocarpine model. Treatment with 150 mg/kg PDTC before and following status epilepticus significantly increased the mortality rate to 100%. Administration of 50 mg/kg PDTC (low-dose) did not exert major effects on the development of a status epilepticus or the mortality rate. In vehicle-treated rats, status epilepticus caused pronounced neuronal damage in the piriform cortex comprising both pyramidal cells and interneurons. Low-dose PDTC treatment almost completely protected from lesions in the piriform cortex. A significant decrease in neuronal density of the hippocampal hilar formation was identified in vehicle- and PDTC-treated rats following status epilepticus. In conclusion, the NF-kappaB inhibitor and antioxidant PDTC protected the piriform cortex, whereas it did not affect hilar neuronal loss. These data might indicate that the generation of reactive oxygen species and activation of NF-kappaB plays a more central role in seizure-associated neuronal damage in the temporal cortex as compared to the hippocampal hilus. However, future investigations are necessary to exactly analyze the biochemical mechanisms by which PDTC exerted its beneficial effects in the piriform cortex.


Subject(s)
Cytoprotection , Neurons/drug effects , Olfactory Pathways/drug effects , Pilocarpine/toxicity , Pyrrolidines/pharmacology , Status Epilepticus/pathology , Thiocarbamates/pharmacology , Animals , Antioxidants/pharmacology , Cell Count , Female , Hippocampus/drug effects , Hippocampus/pathology , NF-kappa B/agonists , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Neurons/pathology , Neuroprotective Agents/pharmacology , Olfactory Pathways/pathology , Rats , Rats, Wistar , Statistics, Nonparametric , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy
8.
J Pharmacol Exp Ther ; 330(3): 939-47, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19494186

ABSTRACT

Up-regulation of the blood-brain barrier efflux transporter P-glycoprotein in central nervous system disorders results in restricted brain access and limited efficacy of therapeutic drugs. In epilepsies, seizure activity strongly triggers expression of P-glycoprotein. Here, we identified the prostaglandin E2 receptor, EP1, as a key factor in the signaling pathway that mediates seizure-induced up-regulation of P-glycoprotein at the blood-brain barrier. In the rat pilocarpine model, status epilepticus significantly increased P-glycoprotein expression by 92 to 197% in the hippocampal hilus and granule cell layer as well as the piriform cortex. The EP1 receptor antagonist 8-chlorodibenz[b,f][1,4]oxazepine-10(11H)-carboxylic acid, 2-[1-oxo-3-(4-pyridinyl)propyl]hydrazide hydrochloride (SC-51089) abolished seizure-induced P-glycoprotein up-regulation and retained its expression at the control level. The control of P-glycoprotein expression despite prolonged seizure activity suggests that EP1 receptor antagonism will also improve antiepileptic drug efficacy. Preliminary evidence for this concept has been obtained using a massive kindling paradigm during which animals received a subchronic SC-51089 treatment. After withdrawal of the EP1 receptor antagonist, a low dose of the P-glycoprotein substrate phenobarbital resulted in an anticonvulsant effect in this pretreated group, whereas the same dosage of phenobarbital did not exert a significant effect in the respective control group. In conclusion, our data demonstrate that EP1 is a key signaling factor in the regulatory pathway that drives P-glycoprotein up-regulation during seizures. These findings suggest new intriguing possibilities to prevent and interrupt P-glycoprotein overexpression in epilepsy. Future studies are necessary to further evaluate the appropriateness of the strategy to enhance the efficacy of antiepileptic drugs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Receptors, Prostaglandin E/drug effects , Seizures/prevention & control , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Anticonvulsants/pharmacology , Blotting, Western , Capillaries/drug effects , Electrodes, Implanted , Female , Image Processing, Computer-Assisted , Immunohistochemistry , Kindling, Neurologic/drug effects , Muscarinic Agonists , Phenobarbital/pharmacology , Pilocarpine , Rats , Rats, Wistar , Receptors, Prostaglandin E, EP1 Subtype , Seizures/genetics , Status Epilepticus/chemically induced , Status Epilepticus/prevention & control , Up-Regulation/physiology
9.
PLoS One ; 3(3): e1813, 2008 Mar 19.
Article in English | MEDLINE | ID: mdl-18350150

ABSTRACT

The thioredoxin-dependent system is an essential regulator of cellular redox balance. Since oxidative stress has been linked with neurodegenerative disease, we studied the roles of thioredoxin reductases in brain using mice with nervous system (NS)-specific deletion of cytosolic (Txnrd1) and mitochondrial (Txnrd2) thioredoxin reductase. While NS-specific Txnrd2 null mice develop normally, mice lacking Txnrd1 in the NS were significantly smaller and displayed ataxia and tremor. A striking patterned cerebellar hypoplasia was observed. Proliferation of the external granular layer (EGL) was strongly reduced and fissure formation and laminar organisation of the cerebellar cortex was impaired in the rostral portion of the cerebellum. Purkinje cells were ectopically located and their dendrites stunted. The Bergmann glial network was disorganized and showed a pronounced reduction in fiber strength. Cerebellar hypoplasia did not result from increased apoptosis, but from decreased proliferation of granule cell precursors within the EGL. Of note, neuron-specific inactivation of Txnrd1 did not result in cerebellar hypoplasia, suggesting a vital role for Txnrd1 in Bergmann glia or neuronal precursor cells.


Subject(s)
Brain/embryology , Thioredoxin-Disulfide Reductase/metabolism , Animals , Cell Proliferation , Growth , Mice , Mice, Knockout , Movement Disorders/enzymology , Movement Disorders/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...