Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Prog Neurobiol ; 206: 102139, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34391810

RESUMEN

Alzheimer's disease is the most common form of dementia characterized by intracellular aggregates of hyperphosphorylated Tau protein and extracellular accumulation of amyloid ß (Aß) peptides. We previously demonstrated that the purinergic receptor P2X7 (P2X7) plays a major role in Aß-mediated neurodegeneration but the relationship between P2X7 and Tau remained overlooked. Such a link was supported by cortical upregulation of P2X7 in patients with various type of frontotemporal lobar degeneration, including mutation in the Tau-coding gene, MAPT, as well as in the brain of a Tauopathy mouse model (THY-Tau22). Subsequent phenotype analysis of P2X7-deficient Tau mice revealed the instrumental impact of this purinergic receptor. Indeed, while P2X7-deficiency had a moderate effect on Tau pathology itself, we observed a significant reduction of microglia activation and of Tau-related inflammatory mediators, particularly CCL4. Importantly, P2X7 deletion ultimately rescued synaptic plasticity and memory impairments of Tau mice. Altogether, the present data support a contributory role of P2X7 dysregulation on processes governing Tau-induced brain anomalies. Due to the convergent role of P2X7 blockade in both Aß and Tau background, P2X7 inhibitors might prove to be ideal candidate drugs to curb the devastating cognitive decline in Alzheimer's disease and Tauopathies.


Asunto(s)
Enfermedad de Alzheimer , Receptores Purinérgicos P2X7/deficiencia , Tauopatías , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides , Animales , Cognición , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Tauopatías/genética , Proteínas tau/genética
2.
J Alzheimers Dis ; 76(4): 1339-1345, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32623401

RESUMEN

BACKGROUND: Masitinib is a selective tyrosine kinase inhibitor that modulates mast cells activity. A previous phase II study reported a cognitive effect of masitinib in patients with Alzheimer's disease. OBJECTIVE: We aimed to shed light on the mode of action of masitinib in Alzheimer's disease. METHODS/RESULTS: We demonstrated here that chronic oral treatment of APPswe/PSEN1dE9 transgenic mice modeling Alzheimer's disease restored normal spatial learning performance while having no impacts on amyloid-ß loads nor on neuroinflammation. However, masitinib promoted a recovery of synaptic markers. Complete genetic depletion of mast cells in APPswe/PSEN1dE9 mice similarly rescued synaptic impairments. CONCLUSION: These results underline that masitinib therapeutic efficacy might primarily be associated with a synapto-protective action in relation with mast cells inhibition.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Cognición/efectos de los fármacos , Sinapsis/efectos de los fármacos , Tiazoles/farmacología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Benzamidas , Modelos Animales de Enfermedad , Masculino , Ratones Transgénicos , Piperidinas , Presenilina-1/genética , Presenilina-1/farmacología , Piridinas , Tiazoles/administración & dosificación
3.
Mol Neurobiol ; 56(8): 5815-5834, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30684218

RESUMEN

Current evidence suggests dementia and pathology in Alzheimer's Disease (AD) are both dependent and independent of amyloid processing and can be induced by multiple 'hits' on vital neuronal functions. Type 2 diabetes (T2D) poses the most important risk factor for developing AD after ageing and dysfunctional IR/PI3K/Akt signalling is a major contributor in both diseases. We developed a model of T2D, coupling subdiabetogenic doses of streptozotocin (STZ) with a human junk food (HJF) diet to more closely mimic the human condition. Over 35 weeks, this induced classic signs of T2D (hyperglycemia and insulin dysfunction) and a modest, but stable deficit in spatial recognition memory, with very little long-term modification of proteins in or associated with IR/PI3K/Akt signalling in CA1 of the hippocampus. Intracerebroventricular infusion of soluble amyloid beta 42 (Aß42) to mimic the early preclinical rise in Aß alone induced a more severe, but short-lasting deficits in memory and deregulation of proteins. Infusion of Aß on the T2D phenotype exacerbated and prolonged the memory deficits over approximately 4 months, and induced more severe aberrant regulation of proteins associated with autophagy, inflammation and glucose uptake from the periphery. A mild form of environmental enrichment transiently rescued memory deficits and could reverse the regulation of some, but not all protein changes. Together, these data identify mechanisms by which T2D could create a modest dysfunctional neuronal milieu via multiple and parallel inputs that permits the development of pathological events identified in AD and memory deficits when Aß levels are transiently effective in the brain.


Asunto(s)
Enfermedad de Alzheimer/epidemiología , Enfermedad de Alzheimer/etiología , Diabetes Mellitus Tipo 2/complicaciones , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/administración & dosificación , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Conducta Alimentaria , Hipocampo/patología , Hipocampo/ultraestructura , Humanos , Insulina/sangre , Masculino , Memoria , Trastornos de la Memoria/complicaciones , Modelos Biológicos , Fosforilación , Ratas Sprague-Dawley , Factores de Riesgo , Estreptozocina , Aumento de Peso
4.
Mol Psychiatry ; 24(1): 108-125, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29934546

RESUMEN

Extracellular aggregates of amyloid ß (Aß) peptides, which are characteristic of Alzheimer's disease (AD), act as an essential trigger for glial cell activation and the release of ATP, leading to the stimulation of purinergic receptors, especially the P2X7 receptor (P2X7R). However, the involvement of P2X7R in the development of AD is still ill-defined regarding the dual properties of this receptor. Particularly, P2X7R activates the NLRP3 inflammasome leading to the release of the pro-inflammatory cytokine, IL-1ß; however, P2X7R also induces cleavage of the amyloid precursor protein generating Aß peptides or the neuroprotective fragment sAPPα. We thus explored in detail the functions of P2X7R in AD transgenic mice. Here, we show that P2X7R deficiency reduced Aß lesions, rescued cognitive deficits and improved synaptic plasticity in AD mice. However, the lack of P2X7R did not significantly affect the release of IL-1ß or the levels of non-amyloidogenic fragment, sAPPα, in AD mice. Instead, our results show that P2X7R plays a critical role in Aß peptide-mediated release of chemokines, particularly CCL3, which is associated with pathogenic CD8+ T cell recruitment. In conclusion, our study highlights a novel detrimental function of P2X7R in chemokine release and supports the notion that P2X7R may be a promising therapeutic target for AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ratones , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
5.
Acta Neuropathol ; 135(6): 839-854, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29696365

RESUMEN

Alzheimer's disease (AD) is associated with a progressive loss of synapses and neurons. Studies in animal models indicate that morphological alterations of dendritic spines precede synapse loss, increasing the proportion of large and short ("stubby") spines. Whether similar alterations occur in human patients, and what their functional consequences could be, is not known. We analyzed biopsies from AD patients and APP x presenilin 1 knock-in mice that were previously shown to present a loss of pyramidal neurons in the CA1 area of the hippocampus. We observed that the proportion of stubby spines and the width of spine necks are inversely correlated with synapse density in frontal cortical biopsies from non-AD and AD patients. In mice, the reduction in the density of synapses in the stratum radiatum was preceded by an alteration of spine morphology, with a reduction of their length and an enlargement of their neck. Serial sectioning examined with electron microscopy allowed us to precisely measure spine parameters. Mathematical modeling indicated that the shortening and widening of the necks should alter the electrical compartmentalization of the spines, leading to reduced postsynaptic potentials in spine heads, but not in soma. Accordingly, there was no alteration in basal synaptic transmission, but long-term potentiation and spatial memory were impaired. These results indicate that an alteration of spine morphology could be involved in the early cognitive deficits associated with AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Espinas Dendríticas/patología , Espinas Dendríticas/fisiología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Simulación por Computador , Modelos Animales de Enfermedad , Femenino , Lóbulo Frontal/patología , Lóbulo Frontal/fisiopatología , Hipocampo/patología , Hipocampo/fisiopatología , Humanos , Imagenología Tridimensional , Masculino , Potenciales de la Membrana/fisiología , Ratones Transgénicos , Microscopía Electrónica , Persona de Mediana Edad , Modelos Neurológicos , Presenilina-1/genética , Presenilina-1/metabolismo , Sinapsis/patología , Técnicas de Cultivo de Tejidos
6.
J Control Release ; 243: 1-10, 2016 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-27671875

RESUMEN

Detection of intracerebral targets with imaging probes is challenging due to the non-permissive nature of blood-brain barrier (BBB). The present work describes two novel single-domain antibodies (VHHs or nanobodies) that specifically recognize extracellular amyloid deposits and intracellular tau neurofibrillary tangles, the two core lesions of Alzheimer's disease (AD). Following intravenous administration in transgenic mouse models of AD, in vivo real-time two-photon microscopy showed gradual extravasation of the VHHs across the BBB, diffusion in the parenchyma and labeling of amyloid deposits and neurofibrillary tangles. Our results demonstrate that VHHs can be used as specific BBB-permeable probes for both extracellular and intracellular brain targets and suggest new avenues for therapeutic and diagnostic applications in neurology.


Asunto(s)
Camélidos del Nuevo Mundo/inmunología , Ovillos Neurofibrilares/inmunología , Placa Amiloide/inmunología , Anticuerpos de Dominio Único/inmunología , Administración Intravenosa , Enfermedad de Alzheimer/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Transgénicos , Microscopía/métodos , Anticuerpos de Dominio Único/metabolismo , Distribución Tisular
7.
Neurobiol Aging ; 36(6): 2043-52, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25862419

RESUMEN

Amyloid-ß (Aß) oligomers are the suspected culprit as initiators of Alzheimer's disease (AD). However, their diffusion in the brain remains unknown. Here, we studied Aß oligomers' dissemination and evaluated their in vivo toxicity. Wild-type mice were injected with 50 pmol of synthetic Aß oligomers (of different size) in the hippocampus. Oligomers diffused largely in the brain as soon as 1 hour and up to 7 days after injection. A transient encephalopathy with memory impairment was induced by this unique injection. The immunoreactivity of the postsynaptic marker PSD95 was diffusely decreased. Similar results (both on memory and PSD95 immunoreactivity) were obtained with delipidated and high molecular weight oligomers (>50 kDa) but not with smaller assemblies. Tau hyperphosphorylation was observed in the oligomer-injected brains. Finally, fos immunostaining was increased in Aß-derived diffusible ligands-injected mice, suggesting neuronal hyperactivity. Rapid and widespread diffusion of Aß oligomers was demonstrated in vivo and associated with decreased synaptic markers and memory deficits which gives new insight to the pathogenicity of Aß.


Asunto(s)
Amnesia/inducido químicamente , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Encefalopatías/inducido químicamente , Enfermedad Aguda , Enfermedad de Alzheimer/etiología , Amnesia/metabolismo , Péptidos beta-Amiloides/administración & dosificación , Animales , Transporte Biológico , Biopolímeros , Encéfalo/metabolismo , Encefalopatías/metabolismo , Difusión , Homólogo 4 de la Proteína Discs Large , Guanilato-Quinasas/metabolismo , Hipocampo , Inyecciones , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Peso Molecular , Fosforilación , Sinapsis/efectos de los fármacos , Factores de Tiempo , Proteínas tau/metabolismo
9.
Nanomedicine ; 9(5): 712-21, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23220328

RESUMEN

Accumulation of amyloid peptide (Aß) in senile plaques is a hallmark lesion of Alzheimer disease (AD). The design of molecules able to target the amyloid pathology in tissue is receiving increasing attention, both for diagnostic and for therapeutic purposes. Curcumin is a fluorescent molecule with high affinity for the Aß peptide but its low solubility limits its clinical use. Curcumin-conjugated nanoliposomes, with curcumin exposed at the surface, were designed. They appeared to be monodisperse and stable. They were non-toxic in vitro, down-regulated the secretion of amyloid peptide and partially prevented Aß-induced toxicity. They strongly labeled Aß deposits in post-mortem brain tissue of AD patients and APPxPS1 mice. Injection in the hippocampus and in the neocortex of these mice showed that curcumin-conjugated nanoliposomes were able to specifically stain the Aß deposits in vivo. Curcumin-conjugated nanoliposomes could find application in the diagnosis and targeted drug delivery in AD. FROM THE CLINICAL EDITOR: In this preclinical study, curcumin-conjugated nanoliposomes were investigated as possible diagnostics and targeted drug delivery system in Alzheimer's disease, demonstrating strong labeling of Aß deposits both in human tissue and in mice, and in vitro downregulation of amyloid peptide secretion and prevention of Aß-induced toxicity.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/química , Curcumina/administración & dosificación , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/aislamiento & purificación , Animales , Autopsia , Colorantes/administración & dosificación , Colorantes/química , Curcumina/química , Humanos , Liposomas/administración & dosificación , Liposomas/química , Ratones , Nanopartículas/administración & dosificación , Nanopartículas/química , Neocórtex/metabolismo , Neocórtex/patología , Fragmentos de Péptidos/química
10.
Neurobiol Aging ; 33(6): 1123.e17-29, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22188721

RESUMEN

Soluble beta-amyloid (Aß) oligomers are considered to putatively play a critical role in the early synapse loss and cognitive impairment observed in Alzheimer's disease. We previously demonstrated that Aß oligomers activate cytosolic phospholipase A(2) (cPLA(2)), which specifically releases arachidonic acid from membrane phospholipids. We here observed that cPLA(2) gene inactivation prevented the alterations of cognitive abilities and the reduction of hippocampal synaptic markers levels noticed upon a single intracerebroventricular injection of Aß oligomers in wild type mice. We further demonstrated that the Aß oligomer-induced sphingomyelinase activation was suppressed and that phosphorylation of Akt/protein kinase B (PKB) was preserved in neuronal cells isolated from cPLA(2)(-/-) mice. Interestingly, expression of the Aß precursor protein (APP) was reduced in hippocampus homogenates and neuronal cells from cPLA(2)(-/-) mice, but the relationship with the resistance of these mice to the Aß oligomer toxicity requires further investigation. These results therefore show that cPLA(2) plays a key role in the Aß oligomer-associated neurodegeneration, and as such represents a potential therapeutic target for the treatment of Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/genética , Enfermedades Neurodegenerativas/enzimología , Fragmentos de Péptidos/toxicidad , Fosfolipasas A2 Citosólicas/fisiología , Animales , Células Cultivadas , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/inducido químicamente , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Enfermedades Neurodegenerativas/inducido químicamente
11.
J Neurosci ; 30(22): 7516-27, 2010 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-20519526

RESUMEN

The development of novel therapeutic strategies for Alzheimer's disease (AD) represents one of the biggest unmet medical needs today. Application of neurotrophic factors able to modulate neuronal survival and synaptic connectivity is a promising therapeutic approach for AD. We aimed to determine whether the loco-regional delivery of ciliary neurotrophic factor (CNTF) could prevent amyloid-beta (Abeta) oligomer-induced synaptic damages and associated cognitive impairments that typify AD. To ensure long-term administration of CNTF in the brain, we used recombinant cells secreting CNTF encapsulated in alginate polymers. The implantation of these bioreactors in the brain of Abeta oligomer-infused mice led to a continuous secretion of recombinant CNTF and was associated with the robust improvement of cognitive performances. Most importantly, CNTF led to full recovery of cognitive functions associated with the stabilization of synaptic protein levels in the Tg2576 AD mouse model. In vitro as well as in vivo, CNTF activated a Janus kinase/signal transducer and activator of transcription-mediated survival pathway that prevented synaptic and neuronal degeneration. These preclinical studies suggest that CNTF and/or CNTF receptor-associated pathways may have AD-modifying activity through protection against progressive Abeta-related memory deficits. Our data also encourage additional exploration of ex vivo gene transfer for the prevention and/or treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Factor Neurotrófico Ciliar/biosíntesis , Factor Neurotrófico Ciliar/uso terapéutico , Trastornos de la Memoria/etiología , Trastornos de la Memoria/terapia , Sinapsis/efectos de los fármacos , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/administración & dosificación , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Apoptosis/genética , Encéfalo/patología , Recuento de Células/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Factor Neurotrófico Ciliar/administración & dosificación , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/farmacología , Transducción de Señal/efectos de los fármacos , Sinapsis/metabolismo , Sinaptosomas/metabolismo , Sinaptosomas/patología , Sinaptosomas/ultraestructura , Factores de Tiempo , Transfección/métodos
12.
Biochimie ; 91(6): 804-9, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19303044

RESUMEN

In the absence of efficient diagnostic and therapeutic tools, Alzheimer's disease (AD) is a major public health concern due to longer life expectancy in the Western countries. Although the precise cause of AD is still unknown, soluble beta-amyloid (Abeta) oligomers are considered the proximate effectors of the synaptic injury and neuronal death occurring in the early stages of AD. Abeta oligomers may directly interact with the synaptic membrane, leading to impairment of synaptic functions and subsequent signalling pathways triggering neurodegeneration. Therefore, membrane structure and lipid status should be considered determinant factors in Abeta-oligomer-induced synaptic and cell injuries, and therefore AD progression. Numerous epidemiological studies have highlighted close relationships between AD incidence and dietary patterns. Among the nutritional factors involved, lipids significantly influence AD pathogenesis. It is likely that maintenance of adequate membrane lipid content could prevent the production of Abeta peptide as well as its deleterious effects upon its interaction with synaptic membrane, thereby protecting neurons from Abeta-induced neurodegeneration. As major constituents of neuronal lipids, n-3 polyunsaturated fatty acids are of particular interest in the prevention of AD valuable diet ingredients whose neuroprotective properties could be essential for designing preventive nutrition-based strategies. In this review, we discuss the functional relevance of neuronal membrane features with respect to susceptibility to Abeta oligomers and AD pathogenesis, as well as the prospective capacities of lipids to prevent or to delay the disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Metabolismo de los Lípidos/fisiología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Apoptosis/fisiología , Ácidos Grasos Omega-3/metabolismo , Humanos , Microdominios de Membrana/metabolismo , Modelos Biológicos , Transducción de Señal/fisiología
13.
Neurobiol Aging ; 29(9): 1319-33, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17459527

RESUMEN

N-terminal-truncated forms of amyloid-beta (A beta) peptide have been recently suggested to play a pivotal role early in Alzheimer's disease (AD). Among them, A beta 3(pE)-42 peptide, starting with pyroglutamyl at residue Glu-3, is considered as the predominant A beta species in AD plaques and pre-amyloid lesions. Its abundance is reported to be directly proportional to the severity of the clinical phenotype. The present study investigates the effects of soluble oligomeric A beta 3(pE)-42 after intracerebroventricular injection on mice learning ability and the molecular mechanisms of its in vitro neurotoxicity. Mice injected with soluble A beta 3(pE)-42 or A beta(l-42) displayed impaired spatial working memory and delayed memory acquisition in Y-maze and Morris water maze tests, while those injected with soluble A beta(42-1) showed no effect. These cognitive alterations were associated with free radical overproduction in the hippocampus and olfactory bulbs, but not in the cerebral cortex or cerebellum. In vitro, A beta 3(pE)-42 oligomers induced a redox-sensitive neuronal apoptosis involving caspase activation and an arachidonic acid-dependent pro-inflammatory pathway. These data suggest that A beta 3(pE)-42 could mediate the neurodegenerative process and subsequent cognitive alteration occurring in preclinical AD stages.


Asunto(s)
Péptidos beta-Amiloides/administración & dosificación , Aprendizaje por Laberinto/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/patología , Fragmentos de Péptidos/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL
14.
Neurobiol Dis ; 23(1): 178-89, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16626961

RESUMEN

Recent data have revealed that soluble oligomeric amyloid-beta peptide (Abeta) may be the proximate effectors of neuronal injuries and death in Alzheimer's disease (AD) by unknown mechanisms. Consistently, we recently demonstrated the critical role of a redox-sensitive cytosolic calcium-dependent phospholipase A2 (cPLA2)-arachidonic acid (AA) pathway in Abeta oligomer-induced cell death. According to the involvement of oxidative stress and polyunsaturated fatty acids like AA in the regulation of sphingomyelinase (SMase) activity, the present study underlines the role of SMases in soluble Abeta-induced apoptosis. Soluble Abeta oligomers induced the activation of both neutral and acidic SMases, as demonstrated by the direct measurement of their enzymatic activities, by the inhibitory effects of both specific neutral and acidic SMase inhibitors, and by gene knockdown using antisense oligonucleotides. Furthermore, soluble Abeta-mediated activation of SMases and subsequent cell death were found to be inhibited by antioxidant molecules and a cPLA2-specific inhibitor or antisense oligonucleotide. We also demonstrate that sphingosine-1-phosphate is a potent neuroprotective factor against soluble Abeta oligomer-induced cell death and apoptosis by inhibiting soluble Abeta-induced activation of acidic sphingomyelinase. These results suggest that Abeta oligomers induce neuronal death by activating neutral and acidic SMases in a redox-sensitive cPLA2-AA pathway.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apoptosis/fisiología , Ceramidas/metabolismo , Neuronas/patología , Fosfolipasas A/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Ácido Araquidónico/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Lisofosfolípidos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oligonucleótidos Antisentido , Oxidación-Reducción , Fosfolipasas A2 , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Esfingosina/análogos & derivados
15.
J Neurochem ; 96(2): 385-95, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16300635

RESUMEN

A growing body of evidence supports the notion that soluble oligomers of amyloid-beta (Abeta) peptide interact with the neuronal plasma membrane, leading to cell injury and inducing death-signalling pathways that could account for the increased neurodegeneration occurring in Alzheimer's disease (AD). Docosahexaenoic acid (DHA, C22:6, n-3) is an essential polyunsaturated fatty acid in the CNS and has been shown in several epidemiological and in vivo studies to have protective effects against AD and cognitive alterations. However, the molecular mechanisms involved remain unknown. We hypothesized that DHA enrichment of plasma membranes could protect neurones from apoptosis induced by soluble Abeta oligomers. DHA pre-treatment was observed to significantly increase neuronal survival upon Abeta treatment by preventing cytoskeleton perturbations, caspase activation and apoptosis, as well as by promoting extracellular signal-related kinase (ERK)-related survival pathways. These data suggest that DHA enrichment probably induces changes in neuronal membrane properties with functional outcomes, thereby increasing protection from soluble Abeta oligomers. Such neuroprotective effects could be of major interest in the prevention of AD and other neurodegenerative diseases.


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/farmacología , Apoptosis/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Neuronas/fisiología , Fármacos Neuroprotectores/farmacología , Animales , Caspasas/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Citoesqueleto/efectos de los fármacos , Citosol/enzimología , Activación Enzimática/efectos de los fármacos , Ácidos Grasos Omega-3/farmacología , Oligodendroglía/fisiología , Estrés Oxidativo/efectos de los fármacos , Fosfolipasas A/metabolismo , Ratas , Ratas Wistar , Solubilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA