Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Pharmacol Rep ; 69(1): 77-80, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27907839

RESUMEN

BACKGROUND: The mechanism of drug resistance in epilepsy remains unknown. Picolinic acid (PIC) is an endogenous metabolite of the kynurenine pathway and a chelating agent added to dietary supplements. Both inhibitory and excitatory properties of PIC were reported. The aim of this study was to determine the influence of exogenously applied PIC upon the electroconvulsive threshold and the activity of chemical convulsants in eight models of epilepsy in mice. METHODS: All experiments were performed on adult male Swiss albino mice. Electroconvulsions were induced through ear clip electrodes. The electroconvulsive threshold (current strength necessary to induce tonic seizures in 50% of the tested group - CS50) was estimated for control animals and animals pretreated with PIC. To determine the possible convulsant activity of PIC, it was administered subcutaneously or intracerebroventricularly in increasing doses to calculate the CD50 values (doses of convulsants necessary to produce seizures in 50% of the animals). Chemical convulsions were induced by challenging the animals with increasing doses of convulsant to calculate the CD50 values. The following convulsants were used: 4-aminopyridine, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, bicuculline, N-methyl-d-aspartate, nicotine, pentylenetrazole, pilocarpine hydrochloride and strychnine nitrate. RESULTS: PIC significantly decreased the electroconvulsive threshold and, after intracerebroventricular injection, but not subcutaneous, produced convulsions. Of the studied convulsants, only the activity of pilocarpine hydrochloride was significantly enhanced by PIC. CONCLUSIONS: PIC enhances seizure activity and potentially may play a role in the pathogenesis of drug resistant epilepsy. Future studies should focus on the interactions between PIC and antiepileptic drugs.


Asunto(s)
Convulsivantes/toxicidad , Electrochoque/efectos adversos , Ácidos Picolínicos/toxicidad , Convulsiones/inducido químicamente , Convulsiones/etiología , Animales , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Electrochoque/métodos , Masculino , Ratones
2.
Pharmacol Rep ; 62(2): 211-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20508276

RESUMEN

Retigabine represents an antiepileptic drug possessing a completely different mechanism of action when compared to the existing classical and newer antiepileptic drugs. In the therapeutic range, retigabine enhances potassium currents, very likely via destabilization of a closed conformation or stabilization of the open conformation of the potassium Kv7.2-7.3 channels. There are also data indicating that this drug may be a GABA enhancer. Kainate-induced status epilepticus in rats resulted in massive apoptosis in the pyriform cortex and hippocampal area - retigabine inhibited neurodegeneration only in the former brain structure. The metabolism of retigabine has nothing to do with cytochrome P450 enzymes and the drug undergoes glucuronidation and acetylation. Randomized, placebo-controlled multicenter studies have shown that retigabine produced a considerable improvement as an add-on drug in patients with partial drug-resistant epilepsy. The most prominent adverse effects due to retigabine combined with the existing antiepileptic treatment were dizziness, somnolence and fatigue. The preclinical data indicate that this antiepileptic drug may possibly be applied in patients with neuropathic pain and affective disorders. Initial clinical data suggest that retigabine may be also effective in Alzheimer's disease or stroke.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Carbamatos/uso terapéutico , Fenilendiaminas/uso terapéutico , Animales , Anticonvulsivantes/farmacocinética , Anticonvulsivantes/farmacología , Carbamatos/farmacología , Ensayos Clínicos como Asunto , Interacciones Farmacológicas , Humanos , Fármacos Neuroprotectores/farmacología , Fenilendiaminas/farmacología
3.
Prog Neuropsychopharmacol Biol Psychiatry ; 34(1): 18-25, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19751793

RESUMEN

The aim of this study was to determine the influence of arachidonyl-2'-chloroethylamide (ACEA - a highly selective cannabinoid type 1 [CB1] receptor agonist) on the protective action and acute adverse effects of carbamazepine, lamotrigine, oxcarbazepine, phenobarbital, phenytoin, and topiramate in the maximal electroshock seizure model and chimney test in mice. Tonic hind limb extension (seizure activity) was evoked in adult male albino Swiss mice by a current (sine-wave, 25 mA, 500 V, 50 Hz, 0.2s stimulus duration) delivered via auricular electrodes. Acute adverse-effect profiles of the studied antiepileptic drugs with respect to motor coordination was assessed in the chimney test. Additionally, long-term memory and skeletal muscular strength were measured along with free plasma (non-protein bound) and total brain antiepileptic drug concentrations. To inhibit the rapid metabolic degradation of ACEA by the fatty-acid amide hydrolase, phenylmethylsulfonyl fluoride (PMSF) was used at a constant ineffective dose of 30 mg/kg. Results indicate that ACEA (2.5 mg/kg, i.p.) co-administered with PMSF (30 mg/kg, i.p.), significantly enhanced the anticonvulsant activity of phenobarbital, but not that of carbamazepine, lamotrigine, oxcarbazepine, phenytoin, or topiramate in the maximal electroshock seizure test in mice. Moreover, ACEA (2.5 mg/kg) with PMSF (30 mg/kg) had no significant impact on the acute adverse effects of all examined antiepileptic drugs in the chimney test in mice. The protective index values (as quotients of the respective TD(50) and ED(50) values denoted from the chimney and maximal electroshock seizure tests, respectively) for the combinations of ACEA (2.5 mg/kg) and PMSF (30 mg/kg) with carbamazepine, oxcarbazepine, phenobarbital, and topiramate were greater than those denoted for the antiepileptic drugs administered alone. Only, the protective index values for the combination of ACEA (2.5 mg/kg) and PMSF (30 mg/kg) with lamotrigine and phenytoin were lower than those determined for the antiepileptic drugs administered alone. Pharmacokinetic experiments revealed that ACEA (2.5 mg/kg) and PMSF (30 mg/kg) affected neither free plasma (non-protein bound) nor total brain concentrations of phenobarbital in mice. Moreover, ACEA and PMSF in combination with carbamazepine, lamotrigine, oxcarbazepine, phenobarbital, phenytoin, and topiramate did not alter long-term memory or skeletal muscular strength in experimental animals. In conclusion, the enhanced anticonvulsant action of phenobarbital by ACEA and PMSF, lack of pharmacokinetic interaction and no acute adverse effects between the examined compounds, make the combination of ACEA and PMSF with phenobarbital of pivotal importance for further experimental and clinical studies. The combinations of ACEA and PMSF with carbamazepine, lamotrigine, oxcarbazepine, phenytoin, and topiramate are neutral from a preclinical viewpoint.


Asunto(s)
Anticonvulsivantes/efectos adversos , Ácidos Araquidónicos/uso terapéutico , Electrochoque/efectos adversos , Receptor Cannabinoide CB1/agonistas , Convulsiones/tratamiento farmacológico , Convulsiones/etiología , Animales , Anticonvulsivantes/metabolismo , Reacción de Prevención/efectos de los fármacos , Modelos Animales de Enfermedad , Combinación de Medicamentos , Inhibidores Enzimáticos/uso terapéutico , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Ratones , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Fluoruro de Fenilmetilsulfonilo/uso terapéutico , Desempeño Psicomotor/efectos de los fármacos , Convulsiones/sangre , Convulsiones/patología
4.
Eur J Pharmacol ; 547(1-3): 65-74, 2006 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-16930590

RESUMEN

Endogenous cannabinoid ligands and cannabinoid CB(1) receptor agonists have been shown to exert potent anticonvulsant effects in various experimental models of epilepsy. The purpose of this study was to determine the effects of arachidonyl-2'-chloroethylamide (ACEA; N-(2-chloroethyl)-5Z,8Z,11Z,14Z-eicosatetraenamide, a highly selective cannabinoid CB(1) receptor agonist) on the threshold for electroconvulsions and the anticonvulsant activity of valproate in the maximal electroshock-induced seizures in mice. To inhibit the rapid metabolic degradation of ACEA by the fatty-acid amide hydrolase, phenylmethylsulfonyl fluoride (PMSF) was used at a constant ineffective dose of 30 mg/kg (i.p.). Moreover, the effects of ACEA and PMSF on the acute adverse-effect profile of valproate were determined in the chimney test. Additionally, the adverse-effect potentials of combination of ACEA, PMSF with valproate were examined in the step-through passive avoidance task (long-term memory) and grip-strength test (neuromuscular strength). To ascertain any pharmacokinetic contribution of ACEA and PMSF to the observed interaction between tested drugs, both free (non-protein bound) plasma and total brain concentrations of valproate were estimated. Results indicated that ACEA (5 and 7.5 mg/kg; i.p.) combined with PMSF increased significantly (P<0.001) the electroconvulsive threshold in mice. ACEA at low doses of 1.25 and 2.5 mg/kg, i.p., with PMSF had no impact on threshold for electroconvulsions. Similarly, neither PMSF (30 mg/kg) nor ACEA (15 mg/kg) administered alone affected the electroconvulsive threshold in mice. Moreover, ACEA (at a subthreshold dose of 2.5 mg/kg; i.p.) co-administered with PMSF potentiated significantly the antielectroshock activity of valproate by reducing its ED(50) from 258.3 to 195.1 mg/kg (P<0.01). Isobolographic transformation of data revealed that the interactions between valproate and ACEA (at 1.25 and 2.5 mg/kg) combined with PMSF were additive. In the chimney test, the combination of ACEA (2.5 mg/kg) and PMSF (30 mg/kg) had no effect on acute adverse effect of valproate and its TD(50) (356.4 mg/kg) did not differ significantly from that for valproate administered alone (TD(50)=404.4 mg/kg). Moreover, none of the examined drugs administered either alone or in combinations produced long-term memory deficits in the step-through passive avoidance task and impaired neuromuscular strength in the grip-strength test in mice. In contrast, ACEA (2.5 mg/kg; i.p.) combined with PMSF (30 mg/kg; i.p.) considerably increased both, the free plasma (by 42%; P<0.01) and total brain (by 49%; P<0.001) concentrations of valproate (administered at 195 mg/kg; i.p.) in mice. Hence, the observed interaction between valproate and ACEA with PMSF in the maximal electroshock test was pharmacokinetic in nature. Finally, based on this preclinical study, one can conclude that ACEA--a cannabinoid CB(1) receptor agonist co-administered with PMSF pharmacokinetically interacted with valproate and thus, providing the enhancement of the antielectroshock activity of valproate in mice, although, the isobolographically determined interaction between drugs was additive. To elucidate the protective role of cannabinoids in the brain during seizures, more advanced neurochemical studies are required.


Asunto(s)
Anticonvulsivantes/farmacología , Ácidos Araquidónicos/farmacología , Receptor Cannabinoide CB1/agonistas , Convulsiones/prevención & control , Ácido Valproico/farmacología , Amidohidrolasas/antagonistas & inhibidores , Amidohidrolasas/metabolismo , Animales , Anticonvulsivantes/sangre , Anticonvulsivantes/farmacocinética , Ácidos Araquidónicos/química , Reacción de Prevención/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Electrochoque/efectos adversos , Inhibidores Enzimáticos/farmacología , Miembro Anterior/efectos de los fármacos , Miembro Anterior/lesiones , Miembro Anterior/fisiología , Masculino , Memoria/efectos de los fármacos , Ratones , Estructura Molecular , Músculos/efectos de los fármacos , Músculos/inervación , Músculos/fisiología , Fluoruro de Fenilmetilsulfonilo/farmacología , Convulsiones/etiología , Convulsiones/fisiopatología , Factores de Tiempo , Ácido Valproico/sangre , Ácido Valproico/farmacocinética
5.
Epilepsia ; 47(1): 10-20, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16417526

RESUMEN

PURPOSE: Approximately 30% of patients with epilepsy do not experience satisfactory seizure control with antiepileptic drug (AED) monotherapy and often require polytherapy. The potential usefulness of AED combinations, in terms of efficacy and adverse effects, is therefore of major importance. The present study sought to identify potentially useful AED combinations with levetiracetam (LEV) METHODS: With isobolographic analysis, the mouse maximal electroshock (MES)-induced seizure model was investigated with regard to the anticonvulsant effects of carbamazepine (CBZ), phenytoin, phenobarbital (PB), valproate, lamotrigine, topiramate (TPM), and oxcarbazepine (OXC), administered singly and in combination with LEV. Acute adverse effects were ascertained by use of the chimney test evaluating motor performance and the step-through passive-avoidance task assessing long-term memory. Brain AED concentrations were determined to ascertain any pharmacokinetic contribution to the observed antiseizure effect. RESULTS: LEV in combination with TPM, at the fixed ratios of 1:2, 1:1, 2:1, and 4:1, was supraadditive (synergistic) in the MES test. Likewise, the combination of LEV with CBZ (at the fixed ratio of 16:1) and LEV with OXC (8:1 and 16:1) were supraadditive. In contrast, all other LEV/AED combinations displayed additivity. Furthermore, none of the investigated LEV/AED combinations altered motor performance and long-term memory. LEV brain concentrations were unaffected by concomitant AED administration, and LEV had no significant effect on brain concentrations of concomitant AEDs. CONCLUSIONS: These preclinical data would suggest that LEV in combination with TPM is associated with beneficial anticonvulsant pharmacodynamic interactions. Similar, but less profound effects were seen with OXC and CBZ.


Asunto(s)
Anticonvulsivantes/farmacología , Anticonvulsivantes/farmacocinética , Modelos Animales de Enfermedad , Piracetam/análogos & derivados , Convulsiones/prevención & control , Animales , Anticonvulsivantes/uso terapéutico , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Carbamazepina/análogos & derivados , Carbamazepina/farmacocinética , Carbamazepina/farmacología , Carbamazepina/uso terapéutico , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Quimioterapia Combinada , Electrochoque , Epilepsia/tratamiento farmacológico , Humanos , Levetiracetam , Masculino , Memoria/efectos de los fármacos , Ratones , Actividad Motora/efectos de los fármacos , Oxcarbazepina , Piracetam/farmacocinética , Piracetam/farmacología , Piracetam/uso terapéutico , Convulsiones/etiología
6.
Eur Neuropsychopharmacol ; 15(6): 609-16, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15922567

RESUMEN

The effect of levetiracetam (LEV) on the acute neurotoxic profiles of various antiepileptic drugs (carbamazepine [CBZ], phenytoin [PHT], phenobarbital [PB], valproate [VPA], lamotrigine [LTG], topiramate [TPM], oxcarbazepine [OXC], and felbamate [FBM]) was evaluated in the rotarod test, allowing the determination of median toxic doses (TD50 values) with respect to impairment of motor coordination in mice. The TD50 of LEV administered singly was 1601 mg/kg. Whilst LEV at 150 mg/kg, being its TID50 (a dose increasing the electroconvulsive threshold by 50%), was without effect with regards to motor coordination impairment associated with PHT, PB, VPA, LTG, OXC, and FBM, it significantly enhanced that associated with CBZ and TPM co-administration. Thus LEV (150 mg/kg) significantly decreased the TD50 of CBZ from 53.6 to 37.3 mg/kg (P<0.01) and that of TPM from 423 to 246 mg/kg (P<0.01). In addition LEV (75 mg/kg) significantly decreased the TD50 of TPM from 423 to 278 (P<0.01). That concurrent measurement of total brain LEV, CBZ, and TPM concentrations showed that concentrations were not significantly different when AEDs were administered singly compared to when they were administered in combination would suggest that there is no pharmacokinetic interaction between these AEDs. Thus, the observed potentialization of the acute neurotoxic effects of CBZ and TPM by LEV is the consequence of a pharmacodynamic interaction. These data support both experimental and clinical published data advocating that LEV may interact with some AEDs by pharmacodynamic mechanisms.


Asunto(s)
Anticonvulsivantes/toxicidad , Carbamazepina/toxicidad , Fructosa/análogos & derivados , Piracetam/análogos & derivados , Equilibrio Postural/efectos de los fármacos , Animales , Anticonvulsivantes/farmacocinética , Encéfalo/metabolismo , Carbamazepina/farmacocinética , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Electrochoque , Fructosa/farmacocinética , Fructosa/toxicidad , Levetiracetam , Masculino , Ratones , Piracetam/farmacocinética , Piracetam/toxicidad , Desempeño Psicomotor/efectos de los fármacos , Topiramato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA