Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nature ; 624(7992): 611-620, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37907096

RESUMEN

Ageing is a critical factor in spinal-cord-associated disorders1, yet the ageing-specific mechanisms underlying this relationship remain poorly understood. Here, to address this knowledge gap, we combined single-nucleus RNA-sequencing analysis with behavioural and neurophysiological analysis in non-human primates (NHPs). We identified motor neuron senescence and neuroinflammation with microglial hyperactivation as intertwined hallmarks of spinal cord ageing. As an underlying mechanism, we identified a neurotoxic microglial state demarcated by elevated expression of CHIT1 (a secreted mammalian chitinase) specific to the aged spinal cords in NHP and human biopsies. In the aged spinal cord, CHIT1-positive microglia preferentially localize around motor neurons, and they have the ability to trigger senescence, partly by activating SMAD signalling. We further validated the driving role of secreted CHIT1 on MN senescence using multimodal experiments both in vivo, using the NHP spinal cord as a model, and in vitro, using a sophisticated system modelling the human motor-neuron-microenvironment interplay. Moreover, we demonstrated that ascorbic acid, a geroprotective compound, counteracted the pro-senescent effect of CHIT1 and mitigated motor neuron senescence in aged monkeys. Our findings provide the single-cell resolution cellular and molecular landscape of the aged primate spinal cord and identify a new biomarker and intervention target for spinal cord degeneration.


Asunto(s)
Senescencia Celular , Quitinasas , Microglía , Neuronas Motoras , Primates , Médula Espinal , Animales , Humanos , Biomarcadores/metabolismo , Quitinasas/metabolismo , Microglía/enzimología , Microglía/metabolismo , Microglía/patología , Neuronas Motoras/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/patología , Primates/metabolismo , Reproducibilidad de los Resultados , Análisis de Expresión Génica de una Sola Célula , Médula Espinal/metabolismo , Médula Espinal/patología
2.
Biochem Biophys Res Commun ; 484(2): 450-455, 2017 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-28137587

RESUMEN

The AMP-activated protein kinase (AMPK) is a key energy sensor. Its activator metformin could suppress epileptogenesis in the pentylenetetrazol (PTZ) kindling model. However, the effect of metformin on the acute and chronic seizures has not been studied. We first detected the expression of AMPK in the brain tissue of human and mice with chronic seizures, as well as in mice with acute seizures. Second, using behavioral assay and local filed potentials (LFPs) recording, we investigated the effect of chronic metformin treatment on seizures in a acute seizure model and a chronic seizure model. Our results showed that AMPK was expressed in neurons in the epileptic brain. The expression level was decreased in the brain tissue that experienced chronic and acute seizures. In PTZ-induced acute seizures model, behavioral assay showed that chronic metformin treatment decreased the mortality, and LFPs recording showed that chronic metformin treatment shortened the duration of generalized tonic-clonic seizures and prolonged the duration of postictal depression. Moreover, in kainic acid-induced chronic seizures model, LFPs recording showed that chronic metformin treatment shortened the duration of epileptic activity. Our study suggests that chronic metformin treatment could facilitate seizure termination.


Asunto(s)
Metformina/farmacología , Convulsiones/prevención & control , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos BALB C , Pentilenotetrazol/toxicidad , Convulsiones/inducido químicamente
3.
Cell Mol Neurobiol ; 37(5): 857-867, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27592227

RESUMEN

SH3 and multiple ankyrin (ANK) repeat domain 3 (SHANK3) is a synaptic scaffolding protein enriched in the postsynaptic density of excitatory synapses. SHANK3 plays an important role in the formation and maturation of excitatory synapses. In the brain, SHANK3 directly or indirectly interacts with various synaptic molecules including N-methyl-D-aspartate receptor, the metabotropic glutamate receptor (mGluR), and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor. Previous studies have shown that Autism spectrum disorder is a result of mutations of the main SHANK3 isoforms, which may be due to deficit in excitatory synaptic transmission and plasticity. Recently, accumulating evidence has demonstrated that overexpression of SHANK3 could induce seizures in vivo. However, little is known about the role of SHANK3 in refractory temporal lobe epilepsy (TLE). Therefore, we investigated the expression pattern of SHANK3 in patients with intractable temporal lobe epilepsy and in pilocarpine-induced models of epilepsy. Immunofluorescence, immunohistochemistry, and western blot analysis were used to locate and determine the expression of SHANK3 in the temporal neocortex of patients with epilepsy, and in the hippocampus and temporal lobe cortex of rats in a pilocarpine-induced epilepsy model. Double-labeled immunofluorescence showed that SHANK3 was mainly expressed in neurons. Western blot analysis confirmed that SHANK3 expression was increased in the neocortex of TLE patients and rats. These results indicate that SHANK3 participates in the pathology of epilepsy.


Asunto(s)
Epilepsia del Lóbulo Temporal/metabolismo , Neocórtex/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Lóbulo Temporal/metabolismo , Adulto , Animales , Conducta Animal , Estudios de Casos y Controles , Demografía , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/patología , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Masculino , Neocórtex/patología , Ratas Sprague-Dawley , Lóbulo Temporal/patología , Adulto Joven
4.
Biochem Biophys Res Commun ; 478(1): 241-246, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27425250

RESUMEN

Glypican-4 (Gpc4) has been found to play an important role in enhancing miniature excitatory postsynaptic currents (mEPSCs). But, the relationship between Gpc4 and epilepsy is still a mystery. In this study, we investigated the expression patterns of Gpc4 in patients with epilepsy and in a pilocarpine-induced rat model of epilepsy. We also determined if altered Gpc4 expression resulted in increased susceptibility to seizures. Western blotting and immunofluorescent methods were utilized. Gpc4 was significantly increased in patients and epileptic rats induced by pilocarpine injection. According to behavioral studies, downregulation of Gpc4 by Gpc4 siRNA decreased spontaneous seizure frequency, while upregulation of Gpc4 by recombinant Gpc4 overexpression led to a converse result. These findings support the hypothesis that increased expression of Gpc4 in the brain is associated with epileptic seizures.


Asunto(s)
Potenciales de Acción , Encéfalo/fisiopatología , Epilepsia/fisiopatología , Glipicanos/metabolismo , Adolescente , Adulto , Animales , Células Cultivadas , Niño , Preescolar , Femenino , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Distribución Tisular , Adulto Joven
5.
Protein Cell ; 15(1): 36-51, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37158785

RESUMEN

Hypoxia-inducible factor (HIF-1α), a core transcription factor responding to changes in cellular oxygen levels, is closely associated with a wide range of physiological and pathological conditions. However, its differential impacts on vascular cell types and molecular programs modulating human vascular homeostasis and regeneration remain largely elusive. Here, we applied CRISPR/Cas9-mediated gene editing of human embryonic stem cells and directed differentiation to generate HIF-1α-deficient human vascular cells including vascular endothelial cells, vascular smooth muscle cells, and mesenchymal stem cells (MSCs), as a platform for discovering cell type-specific hypoxia-induced response mechanisms. Through comparative molecular profiling across cell types under normoxic and hypoxic conditions, we provide insight into the indispensable role of HIF-1α in the promotion of ischemic vascular regeneration. We found human MSCs to be the vascular cell type most susceptible to HIF-1α deficiency, and that transcriptional inactivation of ANKZF1, an effector of HIF-1α, impaired pro-angiogenic processes. Altogether, our findings deepen the understanding of HIF-1α in human angiogenesis and support further explorations of novel therapeutic strategies of vascular regeneration against ischemic damage.


Asunto(s)
Células Endoteliales , Factor A de Crecimiento Endotelial Vascular , Humanos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Endoteliales/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulación de la Expresión Génica , Hipoxia/metabolismo , Hipoxia de la Célula/fisiología
6.
EBioMedicine ; 47: 470-483, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31474551

RESUMEN

BACKGROUND: NACHT and WD repeat domain-containing protein 1 (Nwd1) is a member of the innate immune protein subfamily. Nwd1 contributes to the androgen receptor signaling pathway and is involved in axonal growth. However, the mechanisms that underlie pathophysiological dysfunction in seizures remain unclear. METHODS: Biochemical methods were used to assess Nwd1 expression and localization in a mouse model of kainic acid (KA)-induced acute seizures and temporal lobe epilepsy (TLE) patients. Electrophysiological recordings were used to measure the role of Nwd1 in regulating synaptic transmission and neuronal hyperexcitability in a model of magnesium-free-induced seizure in vitro. Behavioral experiments were performed, and seizure-induced pathological changes were evaluated in a KA-induced seizure model in vivo. GluN2B expression was measured and its correlation with Tyr1472-GluN2B phosphorylation was analyzed in primary hippocampal neurons. FINDINGS: We demonstrated high protein levels of Nwd1 in brain tissues obtained from mice with acute seizures and TLE patients. Silencing Nwd1 in mice using an adeno-associated virus (AAV) profoundly suppressed neuronal hyperexcitability and the occurrence of acute seizures, which may have been caused by reducing GluN2B-containing NMDA receptor-dependent glutamatergic synaptic transmission. Moreover, the decreased activation of Nwd1 reduced GluN2B expression and the phosphorylation of the GluN2B subunit at Tyr1472. INTERPRETATION: Here, we report a previously unrecognized but important role of Nwd1 in seizure models in vitro and in vivo, i.e., modulating the phosphorylation of the GluN2B subunit at Tyr1472 and regulating neuronal hyperexcitability. Meanwhile, our findings may provide a therapeutic strategy for the treatment of epilepsy or other hyperexcitability-related neurological disorders. FUND: The funders have not participated in the study design, data collection, data analysis, interpretation, or writing of the report.


Asunto(s)
Potenciales Evocados/efectos de los fármacos , Hipocampo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ácido Kaínico/efectos adversos , Ratones , Fosforilación , Convulsiones/etiología , Convulsiones/metabolismo , Convulsiones/fisiopatología , Sinapsis/genética , Sinapsis/metabolismo , Transmisión Sináptica
7.
Sci Adv ; 4(10): eaau2357, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30345361

RESUMEN

Epilepsy is a common neurological disease, and approximately 30% of patients do not respond adequately to antiepileptic drug treatment. Recent studies suggest that G protein-coupled receptor 40 (GPR40) is expressed in the central nervous system and is involved in the regulation of neurological function. However, the impact of GPR40 on epileptic seizures remains unclear. In this study, we first reported that GPR40 expression was increased in epileptic brains. In the kainic acid-induced epilepsy model, GPR40 activation after status epilepticus alleviated epileptic activity, whereas GPR40 inhibition showed the opposite effect. In the pentylenetetrazole-induced kindling model, susceptibility to epilepsy was reduced with GPR40 activation and increased with GPR40 inhibition. Whole-cell patch-clamp recordings demonstrated that GPR40 affected N-methyl-d-aspartate (NMDA) receptor-mediated synaptic transmission. Moreover, GPR40 regulated NR2A and NR2B expression on the surface of neurons. In addition, endocytosis of NMDA receptors and binding of GPR40 with NR2A and NR2B can be regulated by GPR40. Together, our findings indicate that GPR40 modulates epileptic seizures, providing a novel antiepileptic target.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsiones/patología , Adolescente , Adulto , Animales , Estudios de Casos y Controles , Células Cultivadas , Niño , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Receptores Acoplados a Proteínas G/genética , Receptores de N-Metil-D-Aspartato/genética , Convulsiones/genética , Convulsiones/metabolismo , Transmisión Sináptica , Adulto Joven
8.
Cell Death Dis ; 9(8): 795, 2018 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-30038264

RESUMEN

Epilepsy is one of the most prevalent and drug-refractory neurological disorders. Zinc finger DHHC-type containing 8 (ZDHHC8) is a putative palmitoyltransferase that is highly expressed in the brain. However, the impact of ZDHHC8 on seizures remains unclear. We aimed to explore the association of ZDHHC8 with epilepsy and investigate its in epileptogenesis in in vivo and in vitro models through behavioral, electrophysiological, and pathological studies. We used kainic acid- and pilocarpine-induced C57BL/6 mice and magnesium-free-induced pyramidal neurons as experimental epileptic models in this study. We first found increased ZDHHC8 expression in the brains of temporal lobe epilepsy (TLE) patients, similar to that observed in chronic epileptic mice, strongly suggesting that ZDHHC8 is correlated with human epilepsy. In the in vitro seizure models, knocking down ZDHHC8 using recombinant adeno-associated virus (rAAV) delayed seizure precipitation and decreased chronic spontaneous recurrent seizures (SRSs) and epileptiform-like discharges, while ZDHHC8 overexpression had the opposite effect. ZDHHC8 levels were consistent with seizure susceptibility in induced mice with SRSs. In an in vitro magnesium-free model, neuronal hyperexcitability and hypersynchrony were reduced in ZDHHC8-knockdown neurons but were increased in ZDHHC8-overexpressing neurons. To further explore the potential mechanisms, we observed that ZDHHC8 had a significant modulatory effect on 2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl) propanoic acid (AMPA) receptor-related excitatory, but not inhibitory, glutamatergic synaptic neurotransmission, further affecting the inward rectification of AMPA currents in acute hippocampal slices in whole-cell recordings. ZDHHC8 facilitated GluA1 trafficking to the neuronal surface in the hippocampus, as shown by immunoprecipitation and Western blotting. These results suggest that ZDHHC8 may promote the generation and propagation of seizures in humans and that knocking down ZDHHC8 might produce anti-epileptogenic effects in drug-resistant epilepsy. Our study provides evidence that may facilitate the development of an alternative approach for the treatment of epilepsy by modulating AMPA/GluA1-mediated neurotransmission.


Asunto(s)
Aciltransferasas/metabolismo , Epilepsia del Lóbulo Temporal/patología , Aciltransferasas/antagonistas & inhibidores , Aciltransferasas/genética , Adolescente , Adulto , Animales , Encéfalo/metabolismo , Preescolar , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Epilepsia/metabolismo , Epilepsia/patología , Epilepsia del Lóbulo Temporal/metabolismo , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores AMPA/metabolismo , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Convulsiones/patología , Transmisión Sináptica , Adulto Joven , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
9.
Oncotarget ; 8(58): 98242-98257, 2017 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-29228686

RESUMEN

Epilepsy is the most prevalent chronic neurological disorder, and its pathological mechanism indicates that an imbalance between excitatory and inhibitory neurotransmission leads to neuronal hyperexcitability. Previous studies have suggested that dl-3n-butylphthalide (NBP) regulates the excitatory neurotransmitter glutamate in the brains of epileptic mice, however, the mechanisms are unknown. We investigated behavioral and electrophysiological factors in rats using NBP. In an in vivo pentylenetetrazole (PTZ)-induced epileptic seizure animal model, NBP decreased the generalized tonic-clonic seizure (GTCS) severity. In an acute hippocampal slice 4-aminopyridine (4-AP) epilepsy model in vitro, NBP decreased the epileptiform activity and miniature excitatory postsynaptic current (mEPSC) amplitude; there was no change in the miniature inhibitory postsynaptic current (mIPSC) amplitude or frequency. This effect suggested changes in excitatory synaptic transmission, which was altered through postsynaptic GluA2-lacking calcium-permeable AMPA receptors (CP-AMPARs). These findings showed that NBP suppressed epileptiform activity in these epilepsy models and provided the first detailed electrophysiological analysis of the impact of NBP in epilepsy models, which may be employed in future experimental or clinical therapies for patients with epilepsy.

10.
Expert Opin Ther Targets ; 21(12): 1083-1094, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29057721

RESUMEN

OBJECTIVES: Plenty of SH3 (POSH) was originally found to be a key regulator of neuronal apoptosis, axon outgrowth, and neuronal migration. However, the role of POSH in epilepsy has not been reported. METHODS: We investigated the expression of POSH in patients with intractable temporal epilepsy (TLE) and in a kainic acid (KA)-induced mouse model, and then we performed behavioral, electrophysiological and biochemical analyses after the lentivirus (LV)-mediated knockdown or overexpression of POSH in the KA-induced model. RESULTS: POSH overexpression shortened the latency of seizure onset, increased the frequency of spontaneous recurrent seizures, and increased the frequency of electrical epileptic discharges, while POSH knockdown had contrasting effects. Whole-cell patch-clamp recordings confirmed that POSH overexpression and knockdown were associated with increased and decreased miniature excitatory postsynaptic currents (mEPSCs) and N-methyl-D-aspartate receptor (NMDAR)-mediated currents, respectively. Finally, co-immunoprecipitation showed that POSH and NMDA receptor subunit 1 (NMDAR1) precipitated with each other, and western blot analysis revealed that the surface expression of NMDAR1 was altered in the hippocampus of epileptic mice. CONCLUSION: These results show that POSH plays a critical role in the progression of epileptic seizures via NMDAR trafficking and suggest that the protein is a novel target for the treatment of human epilepsy.


Asunto(s)
Epilepsia del Lóbulo Temporal/fisiopatología , Terapia Molecular Dirigida , Receptores de N-Metil-D-Aspartato/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Adolescente , Adulto , Animales , Modelos Animales de Enfermedad , Epilepsia/genética , Epilepsia/fisiopatología , Epilepsia/terapia , Epilepsia del Lóbulo Temporal/genética , Epilepsia del Lóbulo Temporal/terapia , Femenino , Técnicas de Silenciamiento del Gen , Hipocampo/patología , Humanos , Ácido Kaínico/toxicidad , Lentivirus/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Ubiquitina-Proteína Ligasas/genética , Adulto Joven
11.
Neuroscience ; 367: 110-120, 2017 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-29111364

RESUMEN

Cluster of differentiation 36 (CD36) belongs to the class B scavenger receptor family. CD36 is a glycoprotein found on the surface of various cell types and has been implicated in the mechanism of numerous central nervous system (CNS) diseases. However, the relationship between CD36 and epilepsy remains unknown. In this study, we aimed to detect the expression of CD36 in two different chronic epileptic mouse models and determine whether CD36 deficiency leads to suppressive neuronal hyperexcitability and decreased susceptibility of epileptic seizures. Here, we found that CD36 was expressed in the neurons and that CD36 expression was significantly elevated in epileptic mice induced by pentylenetetrazol (PTZ) and kainic acid (KA). Behavioral studies revealed that CD36 deletion in mice (CD36-/- mice) resulted in an attenuated progression of chronic epilepsy compared with wild-type (WT) mice. Whole-cell patch-clamp technique exhibited a decreased frequency of action potentials (APs) in the hippocampal slices of CD36-/- mice. In addition, local field potential (LFP) analysis further indicated that CD36 deletion reduced the frequency and duration of epileptiform-like discharges. These results revealed that CD36 deficiency could produce an antiepileptic effect and could provide new insight into antiepileptic treatment.


Asunto(s)
Antígenos CD36/deficiencia , Regulación de la Expresión Génica/genética , Convulsiones/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Análisis de Varianza , Animales , Antígenos CD36/genética , Convulsivantes/toxicidad , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Técnicas In Vitro , Ácido Kaínico/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp , Pentilenotetrazol/toxicidad , Convulsiones/inducido químicamente , Convulsiones/patología
12.
Front Mol Neurosci ; 10: 100, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28439226

RESUMEN

Phosphodiesterase regulates the homeostasis of cAMP and cGMP, which increase the strength of excitatory neural circuits and/or decrease inhibitory synaptic plasticity. Abnormally, synchronized synaptic transmission in the brain leads to seizures. A phosphodiesterase 10A (PDE10A) inhibitor PF-2545920 has recently attracted attention as a potential therapy for neurological and psychiatric disorders. We hypothesized that PF-2545920 plays an important role in status epilepticus (SE) and investigated the underlying mechanisms. PDE10A was primarily located in neurons, and PDE10A expression increased significantly in patients with temporal lobe epilepsy. PF-2545920 enhanced the hyperexcitability of pyramidal neurons in rat CA1, as measured by the frequency of action potentials and miniature excitatory post-synaptic current. GluA1 and NR2A expression also increased significantly in post-synaptic densities, with or without SE in rats treated with PF-2545920. The ratio of p-GluA1/GluA1 increased in the presence of PF-2545920 in groups with SE. Our results suggest that PF-2545920 facilitates seizure activity via the intracellular redistribution of GluA1 and NR2A in the hippocampus. The upregulation of p-GluA1 may play an important role in trafficking GluA1 to post-synaptic densities. The data suggest it would be detrimental to use the drug in seizure patients and might cause neuronal hyperexcitability in non-epileptic individuals.

13.
Expert Rev Neurother ; 16(4): 449-61, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27010915

RESUMEN

Epilepsy is a chronic neurological disease. Although many anti-epileptic drugs (AEDs) have been developed for clinical use, they have no effect on 20-30% of patients and do not generally prevent epileptogenesis. Because of the long development cycle for new AEDs and the high cost, increasing efforts are being made to find anti-epileptic effects among drugs that are already listed for the treatment of other diseases and repurpose them as potential anti-epileptic treatments. Here, we review the progress that has been made in this field as a result of animal and clinical trials of drugs such as rapamycin, everolimus, losartan, celecoxib, bumetanide and other non-epileptic drugs. These drugs can prevent the epileptogenesis, reduce the epileptic pathological changes, and even be used to treat intractable epilepsy. Their mechanisms of action are completely different from those of existing AEDs, prompting researchers to change their perspectives in the search for new AEDs.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia/tratamiento farmacológico , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Humanos , Transducción de Señal/efectos de los fármacos
14.
CNS Drugs ; 30(8): 677-88, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27395404

RESUMEN

BACKGROUND: Seizure following traumatic brain injury (TBI) constitutes a common complication that requires effective prevention to improve the outcome of TBI. Phenytoin has been the only recommended antiepileptic drug (AED) for seizure prophylaxis; however, several shortcomings have affected its use. Intravenous levetiracetam has been available since 2006 and has been increasingly accepted as a seizure prophylaxis for brain injury, mainly due to its favorable pharmacokinetic features and minimal adverse events profile. However, the efficacy and safety of levetiracetam versus phenytoin for seizure prophylaxis following TBI are not well clarified. OBJECTIVE: The aim of this study was to assess the efficacy and safety of levetiracetam versus phenytoin for seizure prophylaxis following TBI. METHODS: We conducted a search of the MEDLINE, EMBASE, and Cochrane library databases to March 2016, and screened original research that included patients with TBI who received levetiracetam. We included randomized controlled trials (RCTs) or controlled observational cohort studies that compared levetiracetam and phenytoin, as well as uncontrolled case series regarding prophylactic levetiracetam following TBI. The outcomes included early or late seizure prophylaxis and safety. The estimates of seizure prophylaxis were pooled using a meta-analysis, and the estimates for the case series were pooled using descriptive statistics. RESULTS: A total of 1614 patients from 11 studies were included in this review, of whom 1285 patients from eight controlled studies (one RCT and seven cohort studies) were included in the meta-analysis. Levetiracetam was not superior to phenytoin with regard to early seizure prophylaxis (risk ratio [RR] 1.10, 95 % confidence interval [CI] 0.64-1.88); the estimate of early seizure incidence was 0.05 (95 % CI 0.02-0.08). Three studies that assessed late seizure did not indicate the superiority of levetiracetam to phenytoin. There were no differences in mortality during hospitalization or after 6 months, or in the number of patients with adverse reactions between levetiracetam and phenytoin. CONCLUSIONS: Levetiracetam does not appear to be superior to phenytoin in efficacy or safety with regard to early or late seizure prophylaxis following TBI; however, no class I evidence was identified. Additional evidence from high-quality studies is required.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Fenitoína/uso terapéutico , Piracetam/análogos & derivados , Convulsiones/tratamiento farmacológico , Anticonvulsivantes/efectos adversos , Estudios de Cohortes , Humanos , Levetiracetam , Estudios Observacionales como Asunto , Fenitoína/efectos adversos , Piracetam/efectos adversos , Piracetam/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto
15.
Expert Rev Neurother ; 15(10): 1113-21, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26416394

RESUMEN

Status epilepticus (SE) is a common, severe neurological disorder, and prolonged seizures in SE may result in irreversible brain damage in association with high disability and mortality rates. Thus, termination of seizures as soon as possible is vital for the successful treatment of this disease. Levetiracetam, a new broad-spectrum anti-epileptic drug, can be used to rapidly and effectively control SE episodes with few side effects. Thus, an understanding of the use of this drug to treat SE will help clinicians to more effectively control SE and improve patient prognosis.


Asunto(s)
Piracetam/análogos & derivados , Estado Epiléptico/tratamiento farmacológico , Anticonvulsivantes/efectos adversos , Anticonvulsivantes/uso terapéutico , Humanos , Levetiracetam , Nootrópicos/efectos adversos , Nootrópicos/uso terapéutico , Piracetam/efectos adversos , Piracetam/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA