Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Neurobiol Dis ; 76: 126-136, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25600211

RESUMEN

As a prominent inflammatory effector of cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2) mediates brain inflammation and injury in many chronic central nervous system (CNS) conditions including seizures and epilepsy, largely through its receptor subtype EP2. However, EP2 receptor activation might also be neuroprotective in models of excitotoxicity and ischemia. These seemingly incongruent observations expose the delicacy of immune and inflammatory signaling in the brain; thus the therapeutic window for quelling neuroinflammation might vary with injury type and target molecule. Here, we identify a therapeutic window for EP2 antagonism to reduce delayed mortality and functional morbidity after status epilepticus (SE) in mice. Importantly, treatment must be delayed relative to SE onset to be effective, a finding that could be explained by the time-course of COX-2 induction after SE and compound pharmacokinetics. A large number of inflammatory mediators were upregulated in hippocampus after SE with COX-2 and IL-1ß temporally leading many others. Thus, EP2 antagonism represents a novel anti-inflammatory strategy to treat SE with a tightly-regulated therapeutic window.


Asunto(s)
Antiinflamatorios/uso terapéutico , Ciclooxigenasa 2/metabolismo , Indoles/administración & dosificación , Indoles/farmacología , Indoles/uso terapéutico , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores , Estado Epiléptico/tratamiento farmacológico , Estado Epiléptico/metabolismo , Animales , Modelos Animales de Enfermedad , Encefalitis/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos C57BL , Pilocarpina , Transducción de Señal/efectos de los fármacos , Estado Epiléptico/mortalidad
2.
Neurobiol Dis ; 70: 74-89, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24952362

RESUMEN

Prostaglandin E2 (PGE2) regulates membrane excitability, synaptic transmission, plasticity, and neuronal survival. The consequences of PGE2 release following seizures has been the subject of much study. Here we demonstrate that the prostaglandin E2 receptor 1 (EP1, or Ptger1) modulates native kainate receptors, a family of ionotropic glutamate receptors widely expressed throughout the central nervous system. Global ablation of the EP1 gene in mice (EP1-KO) had no effect on seizure threshold after kainate injection but reduced the likelihood to enter status epilepticus. EP1-KO mice that did experience typical status epilepticus had reduced hippocampal neurodegeneration and a blunted inflammatory response. Further studies with native prostanoid and kainate receptors in cultured cortical neurons, as well as with recombinant prostanoid and kainate receptors expressed in Xenopus oocytes, demonstrated that EP1 receptor activation potentiates heteromeric but not homomeric kainate receptors via a second messenger cascade involving phospholipase C, calcium and protein kinase C. Three critical GluK5 C-terminal serines underlie the potentiation of the GluK2/GluK5 receptor by EP1 activation. Taken together, these results indicate that EP1 receptor activation during seizures, through a protein kinase C pathway, increases the probability of kainic acid induced status epilepticus, and independently promotes hippocampal neurodegeneration and a broad inflammatory response.


Asunto(s)
Proteína Quinasa C/metabolismo , Receptores de Ácido Kaínico/metabolismo , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Estado Epiléptico/fisiopatología , Animales , Células Cultivadas , Hipocampo/patología , Hipocampo/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroinmunomodulación/fisiología , Oocitos/fisiología , Ratas Sprague-Dawley , Subtipo EP1 de Receptores de Prostaglandina E/genética , Convulsiones/fisiopatología , Transducción de Señal , Estado Epiléptico/patología , Xenopus
3.
Epilepsia ; 55(1): 17-25, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24446952

RESUMEN

Epilepsy is one of the more prevalent neurologic disorders in the world, affecting approximately 50 million people of different ages and backgrounds. Epileptic seizures propagating through both lobes of the forebrain can have permanent debilitating effects on a patient's cognitive and somatosensory brain functions. Epilepsy, defined by the sporadic occurrence of spontaneous recurrent seizures (SRS), is often accompanied by inflammation of the brain. Pronounced increases in the expression of key inflammatory mediators (e.g., interleukin -1ß [IL-1ß], tumor necrosis factor alpha [TNFα], cyclooxygenase-2 [COX-2], and C-X-C motif chemokine 10 [CXCL10]) after seizures may cause secondary damage in the brain and increase the likelihood of repetitive seizures. The COX-2 enzyme is induced rapidly during seizures. The increased level of COX-2 in specific areas of the epileptic brain can help to identify regions of seizure-induced brain inflammation. A good deal of effort has been expended to determine whether COX-2 inhibition might be neuroprotective and represent an adjunct therapeutic strategy along with antiepileptic drugs used to treat epilepsy. However, the effectiveness of COX-2 inhibitors on epilepsy animal models appears to depend on the timing of administration. With all of the effort placed on making use of COX-2 inhibitors as therapeutic agents for the treatment of epilepsy, inflammation, and neurodegenerative diseases there has yet to be a selective and potent COX-2 inhibitor that has shown a clear therapeutic outcome with acceptable side effects.


Asunto(s)
Ciclooxigenasa 2/fisiología , Epilepsia/enzimología , Animales , Anticonvulsivantes/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/enzimología , Inhibidores de la Ciclooxigenasa 2/farmacología , Epilepsia/tratamiento farmacológico , Humanos , Inflamación/enzimología , Enfermedades Neurodegenerativas/enzimología , Convulsiones/tratamiento farmacológico , Convulsiones/enzimología
4.
Neuropharmacology ; 93: 15-27, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25656476

RESUMEN

Exposure to high levels of organophosphorus compounds (OP) can induce status epilepticus (SE) in humans and rodents via acute cholinergic toxicity, leading to neurodegeneration and brain inflammation. Currently there is no treatment to combat the neuropathologies associated with OP exposure. We recently demonstrated that inhibition of the EP2 receptor for PGE2 reduces neuronal injury in mice following pilocarpine-induced SE. Here, we investigated the therapeutic effects of an EP2 inhibitor (TG6-10-1) in a rat model of SE using diisopropyl fluorophosphate (DFP). We tested the hypothesis that EP2 receptor inhibition initiated well after the onset of DFP-induced SE reduces the associated neuropathologies. Adult male Sprague-Dawley rats were injected with pyridostigmine bromide (0.1 mg/kg, sc) and atropine methylbromide (20 mg/kg, sc) followed by DFP (9.5 mg/kg, ip) to induce SE. DFP administration resulted in prolonged upregulation of COX-2. The rats were administered TG6-10-1 or vehicle (ip) at various time points relative to DFP exposure. Treatment with TG6-10-1 or vehicle did not alter the observed behavioral seizures, however six doses of TG6-10-1 starting 80-150 min after the onset of DFP-induced SE significantly reduced neurodegeneration in the hippocampus, blunted the inflammatory cytokine burst, reduced microglial activation and decreased weight loss in the days after status epilepticus. By contrast, astrogliosis was unaffected by EP2 inhibition 4 d after DFP. Transient treatments with the EP2 antagonist 1 h before DFP, or beginning 4 h after DFP, were ineffective. Delayed mortality, which was low (10%) after DFP, was unaffected by TG6-10-1. Thus, selective inhibition of the EP2 receptor within a time window that coincides with the induction of cyclooxygenase-2 by DFP is neuroprotective and accelerates functional recovery of rats.


Asunto(s)
Inhibidores de la Colinesterasa/toxicidad , Isoflurofato/toxicidad , Fármacos Neuroprotectores/uso terapéutico , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Recuperación de la Función/efectos de los fármacos , Estado Epiléptico/inducido químicamente , Acetilcolinesterasa/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Quimiocinas/genética , Quimiocinas/metabolismo , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Gliosis/inducido químicamente , Hipocampo/patología , Indoles/farmacocinética , Indoles/uso terapéutico , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacocinética , Ratas , Ratas Sprague-Dawley , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores , Estado Epiléptico/tratamiento farmacológico , Estado Epiléptico/patología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA