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.
Transl Neurodegener ; 12(1): 1, 2023 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-36624510

RESUMEN

BACKGROUND: Ribosomal protein S6 kinase 1 (S6K1) is a serine-threonine kinase that has two main isoforms: p70S6K (70-kDa isoform) and p85S6K (85-kDa isoform). p70S6K, with its upstream mammalian target of rapamycin (mTOR), has been shown to be involved in learning and memory and participate in the pathophysiology of Alzheimer's disease (AD). However, the function of p85S6K has long been neglected due to its high similarity to p70S6k. The role of p85S6K in learning and memory is still largely unknown. METHODS: We fractionated the postsynaptic densities to illustrate the differential distribution of p85S6K and p70S6K. Coimmunoprecipitation was performed to unveil interactions between p85S6K and the GluA1 subunit of AMPA receptor. The roles of p85S6K in synaptic targeting of GluA1 and learning and memory were evaluated by specific knockdown or overexpression of p85S6K followed by a broad range of methodologies including immunofluorescence, Western blot, in situ proximity ligation assay, morphological staining and behavioral examination. Further, the expression level of p85S6K was measured in brains from AD patients and AD model mice. RESULTS: p85S6K, but not p70S6K, was enriched in the postsynaptic densities. Moreover, knockdown of p85S6K resulted in defective spatial and recognition memory. In addition, p85S6K could interact with the GluA1 subunit of AMPA receptor through synapse-associated protein 97 and A-kinase anchoring protein 79/150. Mechanistic studies demonstrated that p85S6K could directly phosphorylate GluA1 at Ser845 and increase the amount of GluA1 in synapses, thus sustaining synaptic function and spine densities. Moreover, p85S6K was found to be specifically decreased in the synaptosomal compartment in the brains of AD patients and AD mice. Overexpression of p85S6K ameliorated the synaptic deficits and cognitive impairment in transgenic AD model mice. CONCLUSIONS: These results strongly imply a significant role for p85S6K in maintaining synaptic and cognitive function by interacting with GluA1. The findings provide an insight into the rational targeting of p85S6K as a therapeutic potential for AD.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Animales , Ratones , Enfermedad de Alzheimer/genética , Receptores AMPA , Disfunción Cognitiva/genética , Cognición , Ratones Transgénicos , Mamíferos
2.
Neuroscience ; 408: 239-247, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30981860

RESUMEN

M1 muscarinic receptors have long been identified as a potential therapeutic target for the treatment of cognitive impairment in Alzheimer's disease (AD). Our previous study has shown that M1 receptors promote membrane insertion and synaptic delivery of AMPA receptor GluA1 subunit. In this study, we sought to determine whether activation of M1 receptor would rescue the cognitive impairment in AD model mice through modulation of GluA1 subunit. For the mice injected with aggregated ß-amyloid (Aß) fragments to impair learning and memory, activation of M1 receptors could rescue it by reducing the latency to find the platform and spending more time in the target quadrant in the probe test in the Morris water maze. However, such an effect was ablated in mice with Ser845 residue of GluA1 mutated to alanine. Furthermore, the activation of M1 receptors enhanced the expression of GluA1 and its phosphorylation at Ser845 and drove GluA1 to incorporate with PSD95, a postsynaptic marker, in the hippocampi from Aß-injected wild type mice but not from the mutant mice. Moreover, for 9-month-old APP/PS1 transgenic AD model mice, which may resemble the late AD, M1 receptor activation could not improve the cognitive impairment significantly. In addition, the enhancement of GluA1 expression and its phosphorylation at Ser845 were not observed in their hippocampi. Taken together, the study indicated that M1 receptor activation rescued the cognitive deficit through modulating the trafficking of GluA1-containing AMPA receptors and the therapeutics targeting M1 receptors should aim at mild AD or even pre-AD.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Disfunción Cognitiva/tratamiento farmacológico , Agonistas Muscarínicos/farmacología , Fragmentos de Péptidos/farmacología , Receptores AMPA/metabolismo , Aprendizaje Espacial/efectos de los fármacos , Memoria Espacial/efectos de los fármacos , Animales , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones , Agonistas Muscarínicos/uso terapéutico , Fosforilación , Receptor Muscarínico M1/metabolismo
3.
FASEB J ; 33(5): 6622-6631, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30794430

RESUMEN

M1 muscarinic acetylcholine receptors are highly expressed in key areas that control cognition, such as the cortex and hippocampus, representing one potential therapeutic target for cognitive dysfunctions of Alzheimer's disease and schizophrenia. We have reported that M1 receptors facilitate cognition by promoting membrane insertion of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor AMPA receptor subunit 1 (GluA1) through phosphorylation at Ser845. However, the signaling pathway is still unclear. Here we showed that adenylyl cyclase inhibitor 2',5'-dideoxyadenosine and PKA inhibitor KT5720 inhibited enhancement of phosphorylation of Ser845 and membrane insertion of GluA1 induced by M1 receptor activation. Furthermore, PI3K inhibitor LY294002 and protein kinase B (Akt) inhibitor IV blocked the effects of M1 receptors as well. Remarkably, the increase of the activity of PI3K-Akt signaling induced by M1 receptor activation could be abolished by cAMP-PKA inhibitors. Moreover, inhibiting the mammalian target of rapamycin (mTOR) complex 1, an important downstream effector of PI3K-Akt, by short-term application of rapamycin attenuated the effects of M1 receptors on GluA1. Furthermore, such effect was unrelated to possible protein synthesis promoted by mTOR. Taken together, these data demonstrate that M1 receptor activation induces membrane insertion of GluA1 via a signaling linking cAMP-PKA and PI3K-Akt-mTOR pathways but is irrelevant to protein synthesis.-Zhao, L.-X., Ge, Y.-H., Li, J.-B., Xiong, C.-H., Law, P.-Y., Xu, J.-R., Qiu, Y., Chen, H.-Z. M1 muscarinic receptors regulate the phosphorylation of AMPA receptor subunit GluA1 via a signaling pathway linking cAMP-PKA and PI3K-Akt.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Muscarínico M1/metabolismo , Receptores AMPA/metabolismo , Sistemas de Mensajero Secundario/fisiología , Animales , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratas , Ratas Sprague-Dawley
4.
Mater Sci Eng C Mater Biol Appl ; 97: 650-657, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30678952

RESUMEN

Aristolochia and related plants contain nephrotoxins and mutagens in the form of aristolochic acids (AAs). However, there is still lack of a fast and specific method for monitoring AAs in biological samples. Herein, we synthesized a hybrid magnetic mesoporous carbon-molecularly imprinted polymers (MMC@MIPs) as a novel magnetic solid-phase extraction (MSPE) adsorbent for selective recognition of aristolochic acid I and II from rat urine samples. The choline chloride/glycol-based deep eutectic solvent (DES) and indomethacin were used as the eluent and dummy template molecule accordingly. The morphology, structure property and surface groups of the prepared materials were investigated in sequence, and the optimum conditions of the MMC@MIPs-MSPE procedure were also optimized well. Results showed that the proposed method had a relatively satisfactory recovery (86.7-94.3%), with low standard deviation (<4.85%) and acceptable correlation coefficients (0.991-0.996). Overall, this work not only provides an inexpensive and eco-friendly method to fabricate MMC@MIPs, but also develops a highly promising approach for the detection of aristolochic acid I and II in biological samples.


Asunto(s)
Ácidos Aristolóquicos/orina , Magnetismo , Impresión Molecular , Polímeros/química , Animales , Ácidos Aristolóquicos/aislamiento & purificación , Colina/química , Cromatografía Líquida de Alta Presión , Glicoles/química , Porosidad , Ratas , Extracción en Fase Sólida , Solventes/química , Espectrofotometría Ultravioleta
5.
FASEB J ; 32(8): 4247-4257, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29509512

RESUMEN

M1 muscarinic acetylcholine receptors (M1 mAChRs) are the most abundant muscarinic receptors in the hippocampus and have been shown to have procognitive effects. AMPA receptors (AMPARs), an important subtype of ionotropic glutamate receptors, are key components in neurocognitive networks. However, the role of AMPARs in procognitive effects of M1 mAChRs and how M1 mAChRs affect the function of AMPARs remain poorly understood. Here, we found that basal expression of GluA1, a subunit of AMPARs, and its phosphorylation at Ser845 were maintained by M1 mAChR activity. Activation of M1 mAChRs promoted membrane insertion of GluA1, especially to postsynaptic densities. Impairment of hippocampus-dependent learning and memory by antagonism of M1 mAChRs paralleled the reduction of GluA1 expression, and improvement of learning and memory by activation of M1 mAChRs was accompanied by the synaptic insertion of GluA1 and its increased phosphorylation at Ser845. Furthermore, abrogation of phosphorylation of Ser845 residue of GluA1 ablated M1 mAChR-mediated improvement of learning and memory. Taken together, these results show a functional correlation of M1 mAChRs and GluA1 and the essential role of GluA1 in M1 mAChR-mediated cognitive improvement.-Zhao, L.-X., Ge, Y.-H., Xiong, C.-H., Tang, L., Yan, Y.-H., Law, P.-Y., Qiu, Y., Chen, H.-Z. M1 muscarinic receptor facilitates cognitive function by interplay with AMPA receptor GluA1 subunit.


Asunto(s)
Cognición/fisiología , Subunidades de Proteína/metabolismo , Receptor Muscarínico M1/metabolismo , Receptores AMPA/metabolismo , Animales , Emparejamiento Cromosómico/fisiología , Hipocampo/metabolismo , Aprendizaje/fisiología , Masculino , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/metabolismo , Fosforilación/fisiología , Receptores Muscarínicos/metabolismo
6.
CNS Neurosci Ther ; 21(7): 591-8, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26096047

RESUMEN

AIMS: Mechanistic/mammalian target of rapamycin (mTOR) activation by µ-opioid receptor (OPRM1) participates in antinociceptive tolerance, hyperalgesia, and physical dependence. Our previous study also showed that mTOR activation by OPRM1 could attenuate ß amyloid oligomers-induced neurotoxicity. OPRM1 is demonstrated to interact with FK506-binding protein 12 (FKBP12). It is our great interest to investigate whether OPRM1-mediated mTOR signaling is related to receptor-FKBP12 association. METHODS: The activities of mTOR and its downstream effector p70 S6K were measured by immunoblotting their phosphorylation status. The interaction of receptor with mTOR was detected by co-immunoprecipitation and immunofluorescence. RESULTS: OPRM1 activation by morphine-induced time-dependent mTOR activation. PI3K-specific inhibitor LY294002 only blocked the late phase of mTOR activation. However, morphine-induced mTOR activation was totally blocked at all time points in cells expressing FKBP12 association-deficient mutant receptor. FKBP12 knockdown also blocked morphine-induced mTOR activation. Further analysis demonstrated that morphine treatment enhanced the association of receptor with phosphorylated mTOR, whereas decreased association was observed after FKBP12 knockdown, mTOR inhibition or in cells expressing FKBP12 association-deficient mutant. CONCLUSIONS: OPRM1-FKBP12 association played a key role in OPRM1-mediated mTOR activation, which could underlie the mechanisms of multiple physiological and pathological processes. Thus, our findings provide new avenue to modulating these processes.


Asunto(s)
Analgésicos Opioides/farmacología , Morfina/farmacología , Receptores Opioides mu/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteína 1A de Unión a Tacrolimus/metabolismo , Células HEK293 , Humanos , Inmunoprecipitación , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA