Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Neuropharmacology ; 160: 107785, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31541651

RESUMEN

Alzheimer's Disease (AD) is the most prevalent neurodegenerative disorder. Despite advances in the understanding of its pathophysiology, none of the available therapies prevents disease progression. Excess glutamate plays an important role in excitotoxicity by activating ionotropic receptors. However, the mechanisms modulating neuronal cell survival/death via metabotropic glutamate receptors (mGluRs) are not completely understood. Recent data indicates that CDPPB, a positive allosteric modulator of mGluR5, has neuroprotective effects. Thus, this work aimed to investigate CDPPB treatment effects on amyloid-ß (Aß) induced pathological alterations in vitro and in vivo and in a transgenic mouse model of AD (T41 mice). Aß induced cell death in primary cultures of hippocampal neurons, which was prevented by CDPPB. Male C57BL/6 mice underwent stereotaxic surgery for unilateral intra-hippocampal Aß injection, which induced memory deficits, neurodegeneration, neuronal viability reduction and decrease of doublecortin-positive cells, a marker of immature neurons and neuronal proliferation. Treatment with CDPPB for 8 days reversed neurodegeneration and doublecortin-positive cells loss and recovered memory function. Fourteen months old T41 mice presented cognitive deficits, neuronal viability reduction, gliosis and Aß accumulation. Treatment with CDPPB for 28 days increased neuronal viability (32.2% increase in NeuN+ cells) and reduced gliosis in CA1 region (Iba-1+ area by 31.3% and GFAP+ area by 37.5%) in transgenic animals, without inducing hepatotoxicity. However, it did not reverse cognitive deficit. Despite a four-week treatment did not prevent memory loss in aged transgenic mice, CDPPB is protective against Aß stimulus. Therefore, this drug represents a potential candidate for further investigations as AD treatment.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Benzamidas/farmacología , Fármacos Neuroprotectores/farmacología , Pirazoles/farmacología , Receptor del Glutamato Metabotropico 5/efectos de los fármacos , Regulación Alostérica , Péptidos beta-Amiloides/efectos adversos , Animales , Benzamidas/administración & dosificación , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/tratamiento farmacológico , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Fragmentos de Péptidos/efectos adversos , Pirazoles/administración & dosificación , Receptor del Glutamato Metabotropico 5/metabolismo
2.
Epilepsy Behav ; 64(Pt A): 83-89, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27736661

RESUMEN

Temporal lobe epilepsy (TLE) is the most common form of epilepsy in adults. The pilocarpine (PILO) experimental model of TLE portrays behavioral and pathophysiological changes in rodents that are very similar to those found in humans with TLE. However, this model is associated with an unfortunate high mortality rate. Studies have shown that intrahippocampal injection of PILO, while having a much smaller mortality rate, induces status epilepticus (SE) that secondarily leads to TLE. To the best of our knowledge, the present study was the first to evaluate the cognitive and histological alterations 72h after intrahippocampal microinjection of PILO in C57BL/6 mice. Seventy percent of mice developed status epilepticus (SE) after PILO administration, and all animals survived after SE. Seventy-two hours after SE, mice presented memory impairment in both Novel Object Recognition (recognition index - vehicle: 67.57±4.46% vs PILO: 52.33±3.29%) and Contextual Fear Conditioning (freezing time - vehicle: 203±20.43 vs PILO: 107.80±25.17s) tasks. Moreover, using Nissl and NeuN staining, we observed in PILO-treated mice a significant decrease in cell viability and an increase in neuronal loss in all three hippocampal regions analyzed, cornus ammonis (CA) 1, CA3, and dentate gyrus (DG), in comparison with the control group. Additionally, using Iba-1 staining, we observed in PILO-treated mice a significant increase in microglial proliferation in CA1, CA3, and DG of the hippocampus. Therefore, intrahippocampal PILO microinjection is an efficient route to induce SE and acute postictal epileptogenic-like alterations in C57BL/6 mice.


Asunto(s)
Muerte Celular/efectos de los fármacos , Epilepsia del Lóbulo Temporal/inducido químicamente , Gliosis/inducido químicamente , Hipocampo/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Agonistas Muscarínicos/farmacología , Neuronas/efectos de los fármacos , Pilocarpina/farmacología , Estado Epiléptico/inducido químicamente , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Agonistas Muscarínicos/administración & dosificación , Pilocarpina/administración & dosificación
3.
Sci Rep ; 6: 25226, 2016 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-27142962

RESUMEN

Alzheimer's Disease (AD) is a progressive neurodegenerative disease and the main cause of dementia. Substantial evidences indicate that there is over-activation of the PI3K/Akt/mTOR axis in AD. Therefore, the aim of the present study was to investigate the effects of NVP-BEZ235 (BEZ; dactolisib), a dual PI3K/mTOR inhibitor that is under phase I/II clinical trials for the treatment of some types of cancer, in hippocampal neuronal cultures stimulated with amyloid-ß (Aß) 1-42 and in mice injected with Aß 1-42 in the hippocampus. In cell cultures, BEZ reduced neuronal death induced by Aß. BEZ, but not rapamycin, a mTOR inhibitor, or LY294002, a PI3K inhibitor that also inhibits mTOR, reduced the memory impairment induced by Aß. The effect induced by Aß was also prevented in PI3Kγ(-/-) mice. Neuronal death and microgliosis induced by Aß were reduced by BEZ. In addition, the compound increased IL-10 and TNF-α levels in the hippocampus. Finally, BEZ did not change the phosphorylation of Akt and p70s6K, suggesting that the involvement of PI3K and mTOR in the effects induced by BEZ remains controversial. Therefore, BEZ represents a potential strategy to prevent the pathological outcomes induced by Aß and should be investigated in other models of neurodegenerative conditions.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/patología , Imidazoles/administración & dosificación , Trastornos de la Memoria/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Quinolinas/administración & dosificación , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Imidazoles/farmacología , Ratones , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fármacos Neuroprotectores/farmacología , Quinolinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA