Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neurobiol Aging ; 81: 77-87, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31252207

RESUMEN

Huperzine A (HupA) is a kind of Lycopodium alkaloid with potential disease-modifying qualities that has been reported to protect against ß-amyloid (Aß)-mediated mitochondrial damage in Alzheimer's disease. However, the fundamental molecular mechanism underlying the protective action of HupA against Aß-mediated mitochondrial malfunction is not completely understood. Recently, the mitochondrial enzyme amyloid-binding alcohol dehydrogenase (ABAD) protein has been reported to facilitate Aß-induced mitochondrial damage, resulting in mitochondrial malfunction and cell death. Our study found that HupA, but not the acetylcholinesterase inhibitor tacrine, reduced the deposition of Aß and the ABAD level, and further reduced Aß-ABAD complexes, thereby improving cerebral mitochondrial function in APP/PS1 mice. This was accompanied by attenuated reactive oxygen species overload, as well as increases adenosine triphosphate levels. Moreover, HupA decreased the release of cytochrome-c from mitochondria and the level of cleaved caspase-3, thereby increasing dissociated brain cell viability in APP/PS1 mice. Thus, our study demonstrated that a reduction in ABAD was involved in the protective mechanism of HupA on the cerebral mitochondrial function in APP/PS1 mice.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Alcaloides/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Mitocondrias/metabolismo , Fármacos Neuroprotectores/farmacología , Sesquiterpenos/farmacología , Alcaloides/uso terapéutico , Péptidos beta-Amiloides/efectos adversos , Animales , Encéfalo/citología , Supervivencia Celular/efectos de los fármacos , Citocromos c/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratones Transgénicos , Fármacos Neuroprotectores/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Sesquiterpenos/uso terapéutico , Tacrina
2.
Neurochem Res ; 41(6): 1483-95, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26875732

RESUMEN

ß-Amyloid (Aß) can stimulate microglia to release a variety of proinflammatory cytokines and induce neurotoxicity. Nicotine has been reported to inhibit TNF-α, IL-1, and ROS production in microglia. Mitochondrial permeability transition pore (mPTP) plays an important role in neurotoxicity as well. Here, we investigated whether activating the microglial α7-nAChR has a neuroprotective role on neural stem cells (NSCs) and the function of mPTP in NSCs in this process. The expression of α7-nAChR in rat NSCs was detected by immunocytochemistry and RT-PCR. The viability of microglia and NSCs was examined by MTT assay. The mitochondrial membrane potential (ΔΨm) and morphological characteristics of NSCs was measured by JC-1 staining and transmission electron microscopy respectively. The distribution of cytochrome c in the subcellular regions of NSCs was visualized by confocal laser scanning microscopy, and the expression levels of cyclophilin D and cleaved caspase-3 were assayed by western blot. The apoptotic rate of NSCs was measured by flow cytometry. The expression of α7-nAChR was detected in microglial cells, but no expression was found in NSCs. The viability of rat microglial cells and NSCs was not affected by reagents or coculture itself. Aß1-42-mediated microglial activation impaired the morphology and the ΔΨm of mitochondria of NSCs as well as increased cell apoptosis. However, the damage was attenuated when the α7-nAChRs on microglial cells were activated or the mPTPs on NSCs were blocked. Blockade of mPTPs on NSCs and activation of α7-nAChRs on microglia exhibit neuroprotective roles in Aß-induced neurotoxicity of NSCs.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Microglía/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Células-Madre Neurales/metabolismo , Fragmentos de Péptidos/toxicidad , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Microglía/efectos de los fármacos , Poro de Transición de la Permeabilidad Mitocondrial , Células-Madre Neurales/efectos de los fármacos , Subunidades de Proteína/metabolismo , Ratas , Ratas Sprague-Dawley
3.
J Cardiovasc Pharmacol ; 67(2): 182-90, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26448276

RESUMEN

Pulmonary artery hypertension (PAH) is a chronic progressive disease characterized by persistent elevation of pulmonary arterial vascular pressure. The disease severely limits the function of the right ventricle, causing organ failure and finally leading to death. Despite significant advances in pharmacological treatments, PAH remains an incurable disease with high morbidity and mortality. The histopathological change of PAH is featured by remodeling of the pulmonary vascular. Abnormal proliferation of pulmonary artery smooth muscle cells in peripheral vascular is 1 major pathological finding of pulmonary vascular remodeling. Current therapeutics available for PAH primarily aim at inhibiting the pulmonary vasoconstriction and resisting pulmonary vascular remodeling. To date, only some inhibitors targeting proliferative signaling pathways have been used to suppress the proliferation of pulmonary artery smooth muscle cells and reverse pulmonary vascular remodeling. However, because of serious side effects, their clinical use is limited, and more validation is needed before the inhibitors can be transferred into clinical use. This review will focus on signal mechanisms of vascular remodeling in the development of PAH and give an overview of recent advances in research on inhibitors targeting proliferative pathways.


Asunto(s)
Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Transducción de Señal/fisiología , Remodelación Vascular/fisiología , Animales , Daño del ADN/fisiología , Humanos , Mediadores de Inflamación/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología
4.
Int J Mol Med ; 36(6): 1487-96, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26500047

RESUMEN

Microglia (MG)-induced neurotoxicity, a major determinant of Alzheimer's disease, is closely related to the survival of neural stem cells (NSCs). Heat shock protein 75 (Hsp75) has been reported to exert protective effects against environmental stresses; however, whether or not it protects NSCs against MG-derived soluble factor-induced neurotoxicity remains unclear. In the present study, we constructed NSCs that overexpressed human Hsp75 protein and established a co-culture system in order to elucidate the role of Hsp75 in NSC-MG interactions. The results obtained indicated that Hsp75 expression increased after 12 h of soluble factor induction and continued to increase for up to 36 h of treatment. The overexpression of Hsp75 decreased NSC apoptosis and preserved mitochondrial membrane potential. Further experiments revealed that the overexpression of Hsp75 inhibited the formation of cyclophilin D (CypD)-dependent mitochondrial permeability transition pore (mPTP) involvement in neurotoxicity-mediated mitochondrial dysfunction and suppressed the activation of the mitochondrial apoptotic cascade, as demonstrated by the inhibition of the release of cytochrome c (Cytc) and the activation of caspase-3. The findings of this study demonstrate that Hsp75 overexpression prevents the impairment of NSCs induced by MG-derived soluble factors by regulating the opening of mPTP. Thus, Hsp75 warrants further investigation as a potential candidate for protection against neurotoxicity.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Microglía/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Células-Madre Neurales/metabolismo , Péptidos beta-Amiloides/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular , Técnicas de Cocultivo , Peptidil-Prolil Isomerasa F , Ciclofilinas/farmacología , Citocromos c/metabolismo , Citocinas/metabolismo , Citocinas/farmacología , Proteínas HSP90 de Choque Térmico/genética , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Microglía/citología , Microscopía Fluorescente , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Células-Madre Neurales/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fragmentos de Péptidos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Solubilidad
5.
Int J Clin Exp Pathol ; 8(6): 6425-33, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26261518

RESUMEN

OBJECTIVES: This study aims to explore whether Huperzine A (HupA) could protect neural stem cells against amyloid beta-peptide Aß induced apoptosis in a neural stem cells (NSCs) and microglia co-culture system. METHODS: Rat NSCs and microglial cells were isolated, cultured and identified with immunofluorescence Assays (IFA). Co-culture systems of NSCs and microglial cells were employed using Transwell Permeable Supports. The effects of Aß1-42 on NSCs were studied in 4 groups using co-culture systems: NSCs, Aß+NSCs, co-culture and Aß+co-culture groups. Bromodeoxyuridine (BrdU) incorporation and flow cytometry were utilized to assess the differences of proliferation, differentiation and apoptosis of NSCs between the groups. LQ test was performed to assess the amounts of IL-6, TNF-α and MIP-α secreted, and flow cytometry and Western blotting were used to assess apoptosis of NSCs and the expressions of Bcl-2 and Bax in each group. RESULTS: IFA results showed that isolated rat NSCs were nestin-positive and microglial cells were CD11b/c-positive. Among all the groups, the Aß+co-culture group has the lowest BrdU expression level, the lowest MAP2-positive, ChAT-positive cell counts and the highest NSC apoptosis rate. Smaller amounts of IL-6, TNF-α and MIP-α were being secreted by microglial cells in the HupA+Aß+co-culture group compared with those in the Aß+ co-culture group. Also the Bcl-2: Bax ratio was much higher in the HupA+Aß+co-culture group than in the Aß+co-culture group. CONCLUSIONS: HupA inhibits cell apoptosis through restraining microglia's inflammatory response induced by Aß1-42.


Asunto(s)
Alcaloides/farmacología , Péptidos beta-Amiloides/toxicidad , Apoptosis/efectos de los fármacos , Hipocampo/efectos de los fármacos , Microglía/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Comunicación Paracrina/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Sesquiterpenos/farmacología , Animales , Animales Recién Nacidos , Proteínas Reguladoras de la Apoptosis/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Citoprotección , Hipocampo/metabolismo , Hipocampo/patología , Mediadores de Inflamación/metabolismo , Microglía/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neurogénesis/efectos de los fármacos , Ratas Sprague-Dawley , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA