Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Alzheimers Dis ; 62(1): 213-226, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29439327

RESUMEN

BACKGROUND: With current treatments for Alzheimer's disease (AD) only providing temporary symptomatic benefits, disease modifying drugs are urgently required. This approach relies on improved understanding of the early pathophysiology of AD. A new hypothesis has emerged, in which early memory loss is considered a synapse failure caused by soluble amyloid-ß oligomers (Aßo). These small soluble Aßo, which precede the formation of larger fibrillar assemblies, may be the main cause of early AD pathologies. OBJECTIVE: The aim of the current study was to investigate the effect of acute administration of stabilized low-n amyloid-ß1-42 oligomers (Aßo1-42) on cognitive, inflammatory, synaptic, and neuronal markers in the rat. METHODS: Female and male Lister Hooded rats received acute intracerebroventricular (ICV) administration of either vehicle or 5 nmol of Aßo1-42 (10µL). Cognition was assessed in the novel object recognition (NOR) paradigm at different time points. Levels of inflammatory (IL-1ß, IL-6, TNF-α), synaptic (PSD-95, SNAP-25), and neuronal (n-acetylaspartate, parvalbumin-positive cells) markers were investigated in different brain regions (prefrontal and frontal cortex, striatum, dorsal and ventral hippocampus). RESULTS: Acute ICV administration of Aßo1-42 induced robust and enduring NOR deficits. These deficits were reversed by acute administration of donepezil and rolipram but not risperidone. Postmortem analysis revealed an increase in inflammatory markers, a decrease in synaptic markers and parvalbumin containing interneurons in the frontal cortex, with no evidence of widespread neuronal loss. CONCLUSION: Taken together the results suggest that acute administration of soluble low-n Aßo may be a useful model to study the early mechanisms involved in AD and provide us with a platform for testing novel therapeutic approaches that target the early underlying synaptic pathology.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Cognición/fisiología , Inflamación/metabolismo , Trastornos de la Memoria/metabolismo , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Donepezilo/farmacología , Femenino , Inflamación/tratamiento farmacológico , Inflamación/patología , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Nootrópicos/farmacología , Distribución Aleatoria , Ratas , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Risperidona/farmacología , Rolipram/farmacología , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/patología
2.
Mol Neurobiol ; 55(7): 5594-5610, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28983842

RESUMEN

Amyloid precursor protein (APP) is cleaved not only to generate the amyloid peptide (Aß), involved in neurodegenerative processes, but can also be metabolized by alpha secretase to produce and release soluble N-terminal APP (sAPPα), which has many properties including the induction of axonal elongation and neuroprotection. The mechanisms underlying the properties of sAPPα are not known. Here, we used proteomic analysis of mouse cortico-hippocampal membranes to identify the neuronal specific alpha3 (α3)-subunit of the plasma membrane enzyme Na, K-ATPase (NKA) as a new binding partner of sAPPα. We showed that sAPPα recruits very rapidly clusters of α3-NKA at neuronal surface, and its binding triggers a cascade of events promoting sAPPα-induced axonal outgrowth. The binding of sAPPα with α3-NKA was not observed for sAPPα-induced Aß1-42 oligomers neuroprotection, neither the downstream events particularly the interaction of sAPPα with APP before endocytosis, ERK signaling, and the translocation of SET from the nucleus to the plasma membrane. These data suggest that the mechanisms of the axonal growth promoting and neuroprotective properties of sAPPα appear to be specific and independent. The signals at the cell surface specific to trigger these mechanisms require further study.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Axones/metabolismo , Neuroprotección , Fragmentos de Péptidos/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Membrana Celular/metabolismo , Proteínas de Unión al ADN , Endocitosis , Chaperonas de Histonas , Humanos , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL , Modelos Biológicos , Neuritas/metabolismo , Proteínas Oncogénicas/metabolismo , Unión Proteica , Solubilidad
3.
Alzheimers Res Ther ; 9(1): 69, 2017 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-28851448

RESUMEN

BACKGROUND: Polyunsaturated fatty acids play a crucial role in neuronal function, and the modification of these compounds in the brain could have an impact on neurodegenerative diseases such as Alzheimer's disease. Despite the fact that arachidonic acid is the second foremost polyunsaturated fatty acid besides docosahexaenoic acid, its role and the regulation of its transfer and mobilization in the brain are poorly known. METHODS: Two groups of 39 adult male BALB/c mice were fed with an arachidonic acid-enriched diet or an oleic acid-enriched diet, respectively, for 12 weeks. After 10 weeks on the diet, mice received intracerebroventricular injections of either NaCl solution or amyloid-ß peptide (Aß) oligomers. Y-maze and Morris water maze tests were used to evaluate short- and long-term memory. At 12 weeks on the diet, mice were killed, and blood, liver, and brain samples were collected for lipid and protein analyses. RESULTS: We found that the administration of an arachidonic acid-enriched diet for 12 weeks induced short-term memory impairment and increased deleterious effects of Aß oligomers on learning abilities. These cognitive alterations were associated with modifications of expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, postsynaptic density protein 95, and glial fibrillary acidic protein in mouse cortex or hippocampus by the arachidonic acid-enriched diet and Aß oligomer administration. This diet also led to an imbalance between the main ω-6 fatty acids and the ω-3 fatty acids in favor of the first one in erythrocytes and the liver as well as in the hippocampal and cortical brain structures. In the cortex, the dietary arachidonic acid also induced an increase of arachidonic acid-containing phospholipid species in phosphatidylserine class, whereas intracerebroventricular injections modified several arachidonic acid- and docosahexaenoic acid-containing species in the four phospholipid classes. Finally, we observed that dietary arachidonic acid decreased the expression of the neuronal form of acyl-coenzyme A synthetase 4 in the hippocampus and increased the cytosolic phospholipase A2 activation level in the cortices of the mice. CONCLUSIONS: Dietary arachidonic acid could amplify Aß oligomer neurotoxicity. Its consumption could constitute a risk factor for Alzheimer's disease in humans and should be taken into account in future preventive strategies. Its deleterious effect on cognitive capacity could be linked to the balance between arachidonic acid-mobilizing enzymes.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Ácido Araquidónico/efectos adversos , Dieta/efectos adversos , Aprendizaje por Laberinto/fisiología , Fragmentos de Péptidos/toxicidad , Receptores AMPA/metabolismo , Animales , Ácido Araquidónico/administración & dosificación , Astrocitos/metabolismo , Proliferación Celular/fisiología , Corteza Cerebral/metabolismo , Coenzima A Ligasas/metabolismo , Citosol/metabolismo , Homólogo 4 de la Proteína Discs Large/metabolismo , Hipocampo/metabolismo , Masculino , Trastornos de la Memoria/etiología , Trastornos de la Memoria/fisiopatología , Ratones Endogámicos BALB C , Neuronas/metabolismo , Ácido Oléico/administración & dosificación , Fosfolipasas A2/metabolismo , Factores de Riesgo
4.
J Biol Chem ; 292(30): 12713-12724, 2017 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-28623233

RESUMEN

Alzheimer disease is associated with deposition of the amyloidogenic peptide Aß in the brain. Passive immunization using Aß-specific antibodies has been demonstrated to reduce amyloid deposition both in vitro and in vivo Because N-terminally truncated pyroglutamate (pE)-modified Aß species (AßpE3) exhibit enhanced aggregation potential and propensity to form toxic oligomers, they represent particularly attractive targets for antibody therapy. Here we present three separate monoclonal antibodies that specifically recognize AßpE3 with affinities of 1-10 nm and inhibit AßpE3 fibril formation in vitro. In vivo application of one of these resulted in improved memory in AßpE3 oligomer-treated mice. Crystal structures of Fab-AßpE3 complexes revealed two distinct binding modes for the peptide. Juxtaposition of pyroglutamate pE3 and the F4 side chain (the "pEF head") confers a pronounced bulky hydrophobic nature to the AßpE3 N terminus that might explain the enhanced aggregation properties of the modified peptide. The deep burial of the pEF head by two of the antibodies explains their high target specificity and low cross-reactivity, making them promising candidates for the development of clinical antibodies.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/inmunología , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Inmunoterapia , Ácido Pirrolidona Carboxílico/inmunología , Péptidos beta-Amiloides/química , Animales , Células Cultivadas , Ratones
5.
Nat Commun ; 7: 12504, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-27509875

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) inhibit cyclooxygenase-1 (COX-1) and COX-2 enzymes. The NLRP3 inflammasome is a multi-protein complex responsible for the processing of the proinflammatory cytokine interleukin-1ß and is implicated in many inflammatory diseases. Here we show that several clinically approved and widely used NSAIDs of the fenamate class are effective and selective inhibitors of the NLRP3 inflammasome via inhibition of the volume-regulated anion channel in macrophages, independently of COX enzymes. Flufenamic acid and mefenamic acid are efficacious in NLRP3-dependent rodent models of inflammation in air pouch and peritoneum. We also show therapeutic effects of fenamates using a model of amyloid beta induced memory loss and a transgenic mouse model of Alzheimer's disease. These data suggest that fenamate NSAIDs could be repurposed as NLRP3 inflammasome inhibitors and Alzheimer's disease therapeutics.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Antiinflamatorios no Esteroideos/farmacología , Ácido Flufenámico/farmacología , Inflamasomas/metabolismo , Ácido Mefenámico/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Muerte Celular , Canales de Cloruro/metabolismo , Cisteína/metabolismo , Femenino , Genotipo , Inflamación , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Trastornos de la Memoria/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Reconocimiento Visual de Modelos/efectos de los fármacos , Ratas
6.
Sci Rep ; 5: 17338, 2015 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-26626428

RESUMEN

Full-length Aß1-42 and Aß1-40, N-truncated pyroglutamate Aß3-42 and Aß4-42 are major variants in the Alzheimer brain. Aß4-42 has not been considered as a therapeutic target yet. We demonstrate that the antibody NT4X and its Fab fragment reacting with both the free N-terminus of Aß4-x and pyroglutamate Aß3-X mitigated neuron loss in Tg4-42 mice expressing Aß4-42 and completely rescued spatial reference memory deficits after passive immunization. NT4X and its Fab fragment also rescued working memory deficits in wild type mice induced by intraventricular injection of Aß4-42. NT4X reduced pyroglutamate Aß3-x, Aßx-40 and Thioflavin-S positive plaque load after passive immunization of 5XFAD mice. Aß1-x and Aßx-42 plaque deposits were unchanged. Importantly, for the first time, we demonstrate that passive immunization using the antibody NT4X is therapeutically beneficial in Alzheimer mouse models showing that N-truncated Aß starting with position four in addition to pyroglutamate Aß3-x is a relevant target to fight Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/inmunología , Anticuerpos Monoclonales de Origen Murino/farmacología , Inmunización Pasiva/métodos , Fragmentos de Péptidos/inmunología , Enfermedad de Alzheimer/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino/inmunología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratas
7.
J Alzheimers Dis ; 45(1): 195-204, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25690661

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disease that has been linked to changes in cholesterol metabolism. Neuronal cholesterol content significantly influences the pro-apoptotic effect of amyloid-ß peptide42 (Aß42), which plays a key role in AD development. We previously reported that aged mice with reduced expression of the lipolysis stimulated lipoprotein receptor (LSR+/-), demonstrate membrane cholesterol accumulation and decreased intracellular lipid droplets in several brain regions, suggesting a potential role of LSR in brain cholesterol distribution. We questioned if these changes rendered the LSR+/- mouse more susceptible to Aß42-induced cognitive and biochemical changes. Results revealed that intracerebroventricular injection of oligomeric Aß42 in male 15-month old LSR+/+ and LSR+/- mice led to impairment in learning and long-term memory and decreased cortical cholesterol content of both groups; these effects were significantly amplified in the Aß42-injected LSR+/- group. Total latency of the Morris test was significantly and negatively correlated with cortical cholesterol content of the LSR+/- mice, but not of controls. Significantly lower cortical PSD95 and SNAP-25 levels were detected in Aß42-injected LSR+/- mice as compared to Aß42-injected LSR+/+ mice. In addition, 24S-hydroxy cholesterol metabolite levels were significantly higher in the cortex of LSR+/- mice. Taken together, these results suggest that changes in cortex cholesterol regulation as a result of the LSR+/- genotype were linked to increased susceptibility to amyloid stress, and we would therefore propose the aged LSR+/- mouse as a new model for understanding the link between modified cholesterol regulation as a risk factor for AD.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Colesterol/metabolismo , Fragmentos de Péptidos/farmacología , Receptores de Lipoproteína/deficiencia , Análisis de Varianza , Animales , Homólogo 4 de la Proteína Discs Large , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Guanilato-Quinasas/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hidroxicolesteroles/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero , Receptores de Lipoproteína/genética , Análisis de Regresión , Proteína 25 Asociada a Sinaptosomas/metabolismo
8.
Acta Neuropathol Commun ; 1: 56, 2013 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-24252153

RESUMEN

BACKGROUND: The amyloid hypothesis in Alzheimer disease (AD) considers amyloid ß peptide (Aß) deposition causative in triggering down-stream events like neurofibrillary tangles, cell loss, vascular damage and memory decline. In the past years N-truncated Aß peptides especially N-truncated pyroglutamate AßpE3-42 have been extensively studied. Together with full-length Aß1-42 and Aß1-40, N-truncated AßpE3-42 and Aß4-42 are major variants in AD brain. Although Aß4-42 has been known for a much longer time, there is a lack of studies addressing the question whether AßpE3-42 or Aß4-42 may precede the other in Alzheimer's disease pathology. RESULTS: Using different Aß antibodies specific for the different N-termini of N-truncated Aß, we discovered that Aß4-x preceded AßpE3-x intraneuronal accumulation in a transgenic mouse model for AD prior to plaque formation. The novel Aß4-x immunoreactive antibody NT4X-167 detected high molecular weight aggregates derived from N-truncated Aß species. While NT4X-167 significantly rescued Aß4-42 toxicity in vitro no beneficial effect was observed against Aß1-42 or AßpE3-42 toxicity. Phenylalanine at position four of Aß was imperative for antibody binding, because its replacement with alanine or proline completely prevented binding. Although amyloid plaques were observed using NT4X-167 in 5XFAD transgenic mice, it barely reacted with plaques in the brain of sporadic AD patients and familial cases with the Arctic, Swedish and the presenilin-1 PS1Δ9 mutation. A consistent staining was observed in blood vessels in all AD cases with cerebral amyloid angiopathy. There was no cross-reactivity with other aggregates typical for other common neurodegenerative diseases showing that NT4X-167 staining is specific for AD. CONCLUSIONS: Aß4-x precedes AßpE3-x in the well accepted 5XFAD AD mouse model underlining the significance of N-truncated species in AD pathology. NT4X-167 therefore is the first antibody reacting with Aß4-x and represents a novel tool in Alzheimer research.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Anticuerpos Monoclonales/administración & dosificación , Neuronas/fisiología , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Mapeo Epitopo , Femenino , Humanos , Masculino , Ratones Transgénicos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Placa Amiloide/tratamiento farmacológico , Placa Amiloide/fisiopatología , Ratas Wistar
9.
Acta Neuropathol ; 126(2): 189-205, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23685882

RESUMEN

N-truncated Aß4-42 is highly abundant in Alzheimer disease (AD) brain and was the first Aß peptide discovered in AD plaques. However, a possible role in AD aetiology has largely been neglected. In the present report, we demonstrate that Aß4-42 rapidly forms aggregates possessing a high aggregation propensity in terms of monomer consumption and oligomer formation. Short-term treatment of primary cortical neurons indicated that Aß4-42 is as toxic as pyroglutamate Aß3-42 and Aß1-42. In line with these findings, treatment of wildtype mice using intraventricular Aß injection induced significant working memory deficits with Aß4-42, pyroglutamate Aß3-42 and Aß1-42. Transgenic mice expressing Aß4-42 (Tg4-42 transgenic line) developed a massive CA1 pyramidal neuron loss in the hippocampus. The hippocampus-specific expression of Aß4-42 correlates well with age-dependent spatial reference memory deficits assessed by the Morris water maze test. Our findings indicate that N-truncated Aß4-42 triggers acute and long-lasting behavioral deficits comparable to AD typical memory dysfunction.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/toxicidad , Degeneración Nerviosa/patología , Neuronas/efectos de los fármacos , Enfermedad de Alzheimer/etiología , Amiloide/química , Amiloide/ultraestructura , Péptidos beta-Amiloides/química , Animales , Conducta Animal/fisiología , Femenino , Feto/citología , Humanos , Inyecciones Intraventriculares , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Peso Molecular , Degeneración Nerviosa/etiología , Neuronas/citología , Neuronas/patología , Embarazo , Cultivo Primario de Células , Ratas , Ratas Wistar , Solubilidad , Percepción Espacial/fisiología
10.
J Neurochem ; 123(4): 467-76, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22909011

RESUMEN

Brain lipid homeostasis is important for maintenance of brain cell function and synaptic communications, and is intimately linked to age-related cognitive decline. Because of the blood-brain barrier's limiting nature, this tissue relies on a complex system for the synthesis and receptor-mediated uptake of lipids between the different networks of neurons and glial cells. Using immunofluorescence, we describe the region-specific expression of the lipolysis-stimulated lipoprotein receptor (LSR), in the mouse hippocampus, cerebellum Purkinje cells, the ependymal cell interface between brain parenchyma and cerebrospinal fluid, and the choroid plexus. Colocalization with cell-specific markers revealed that LSR was expressed in neurons, but not astrocytes. Latency in arms of the Y-maze exhibited by young heterozygote LSR(+/-) mice was significantly different as compared to control LSR(+/+), and increased in older LSR(+/-) mice. Filipin and Nile red staining revealed membrane cholesterol content accumulation accompanied by significantly altered distribution of LSR in the membrane, and decreased intracellular lipid droplets in the cerebellum and hippocampus of old LSR(+/-) mice, as compared to control littermates as well as young LSR(+/-) animals. These data therefore suggest a potential role of LSR in brain cholesterol distribution, which is particularly important in preserving neuronal integrity and thereby cognitive functions during aging.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Colesterol/metabolismo , Neuronas/metabolismo , Receptores de LDL/metabolismo , Factores de Edad , Animales , Plexo Coroideo/metabolismo , Filipina/metabolismo , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfopiruvato Hidratasa/metabolismo , Receptores de LDL/deficiencia
11.
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
12.
FASEB J ; 24(11): 4218-28, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20647547

RESUMEN

As a hepatic receptor for triglyceride-rich lipoproteins, the lipolysis-stimulated lipoprotein receptor (LSR) may be involved in the dynamics of lipid distribution between the liver and peripheral tissues. Here, we explore the potential role of leptin in regulating LSR. At physiological concentrations (1-10 ng/ml), leptin increased LSR protein and mRNA levels in Hepa1-6 cells through an ERK1/2-dependent and α-amanitin-sensitive pathway. In vivo, leptin treatment of C57BL6/Rj mice (1 µg 2×/d, 8 d) led to a significant increase in hepatic LSR mRNA and protein, decreased liver triglycerides and increased VLDL secretion as compared to controls. LSR(+/-) mice with elevated postprandial lipemia placed on a high-fat (60% kcal) diet exhibited accelerated weight gain and increased fat mass as compared to controls. While plasma leptin levels were increased 3-fold, hepatic leptin receptor protein levels and phosphorylation of ERK1/2 were significantly reduced. Therefore, leptin is an important regulator of LSR protein levels providing the means for the control of hepatic uptake of lipids during the postprandial phase. However, this may no longer be functional in LSR(+/-) mice placed under a chronic dietary fat load, suggesting that this animal model could be useful for the study of molecular mechanisms involved in peripheral leptin resistance.


Asunto(s)
Leptina/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Lipólisis/efectos de los fármacos , Hígado/efectos de los fármacos , Periodo Posprandial , Receptores de Lipoproteína/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Western Blotting , Peso Corporal/efectos de los fármacos , Línea Celular , Técnica del Anticuerpo Fluorescente , Leptina/sangre , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa
13.
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
14.
Biochim Biophys Acta ; 1801(8): 791-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20211757

RESUMEN

Alzheimer's disease (AD) is a major public health concern due to longer life expectancy in the Western countries. Amyloid-beta (Abeta) oligomers are considered the proximate effectors in the early stages of AD. AD-related cognitive impairment, synaptic loss and neurodegeneration result from interactions of Abeta oligomers with the synaptic membrane and subsequent activation of pro-apoptotic signalling pathways. Therefore, membrane structure and lipid status appear determinant in Abeta-induced toxicity. Numerous epidemiological studies have highlighted the beneficial influence of docosahexaenoic acid (DHA, C22:6 n-3) on the preservation of synaptic function and memory capacities in aged individuals or upon Abeta exposure, whereas its deficiency is presented as a risk factor for AD. An elevated number of studies have been reporting the beneficial effects of dietary DHA supplementation on cognition and synaptic integrity in various AD models. In this review, we describe the important potential of DHA to preserve neuronal and brain functions and classified its numerous molecular and cellular effects from impact on membrane lipid content and organisation to activation of signalling pathways sustaining synaptic function and neuronal survival. DHA appears as one of the most valuable diet ingredients whose neuroprotective properties could be crucial for designing nutrition-based strategies able to prevent AD as well as other lipid- and age-related diseases whose prevalence is progressing in elderly populations.


Asunto(s)
Enfermedad de Alzheimer/patología , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/farmacología , Ratones , Sinapsis/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Animales , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/fisiopatología , Trastornos del Conocimiento/prevención & control , Citoprotección/efectos de los fármacos , Ácidos Docosahexaenoicos/metabolismo , Humanos , Modelos Biológicos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología
15.
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
16.
J Lipid Res ; 49(4): 856-69, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18160739

RESUMEN

The H2 allele of APOC1, giving rise to increased gene expression of apolipoprotein C-I (apoC-I), is in genetic disequilibrium with the APOE4 allele and may provide a major risk factor for Alzheimer's disease (AD). We found that apoC-I protein is present in astrocytes and endothelial cells within hippocampal regions in both human control and AD brains. Interestingly, apoC-I colocalized with beta-amyloid (Abeta) in plaques in AD brains, and in vitro experiments revealed that aggregation of Abeta was delayed in the presence of apoC-I. Moreover, apoC-I was found to exacerbate the soluble Abeta oligomer-induced neuronal death. To establish a potential role for apoC-I in cognitive functions, we used human (h) APOC1(+/0) transgenic mice that express APOC1 mRNA throughout their brains and apoC-I protein in astrocytes and endothelial cells. The hAPOC1(+/0) mice displayed impaired hippocampal-dependent learning and memory functions compared with their wild-type littermates, as judged from their performance in the object recognition task (P = 0.012) and in the Morris water maze task (P = 0.010). ApoC-I may affect learning as a result of its inhibitory properties toward apoE-dependent lipid metabolism. However, no differences in brain mRNA or protein levels of endogenous apoE were detected between transgenic and wild-type mice. In conclusion, human apoC-I expression impairs cognitive functions in mice independent of apoE expression, which supports the potential of a modulatory role for apoC-I during the development of AD.


Asunto(s)
Apolipoproteína C-I/metabolismo , Regulación de la Expresión Génica , Aprendizaje , Memoria , Animales , Apolipoproteína C-I/genética , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/genética , Técnicas de Cultivo de Tejidos
17.
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
18.
J Neurol Sci ; 262(1-2): 27-36, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17681547

RESUMEN

Alzheimer's disease (AD) is a major public health concern in all countries. Although the precise cause of AD is still unknown, a growing body of evidence supports the notion that soluble amyloid beta-peptide (Abeta) may be the proximate cause of synaptic injuries and neuronal death early in the disease. AD patients display lower levels of docosahexaenoic acid (DHA, C22:6 ; n-3) in plasma and brain tissues as compared to age-matched controls. Furthermore, epidemiological studies suggest that high DHA intake might have protective properties against neurodegenerative diseases. These observations are supported by in vivo studies showing that DHA-rich diets limits the synaptic loss and cognitive defects induced by Abeta peptide. Although the molecular basis of these neuroprotective effects remains unknown, several mechanisms have been proposed such as (i) regulation of the expression of potentially protective genes, (ii) activation of anti-inflammatory pathways, (iii) modulation of functional properties of the synaptic membranes along with changes in their physicochemical and structural features.


Asunto(s)
Enfermedad de Alzheimer/dietoterapia , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/antagonistas & inhibidores , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ácidos Docosahexaenoicos/uso terapéutico , Encefalitis/dietoterapia , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/fisiopatología , Ácidos Docosahexaenoicos/metabolismo , Encefalitis/metabolismo , Encefalitis/fisiopatología , Alimentos Formulados/normas , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/fisiología , Fármacos Neuroprotectores/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo
19.
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
20.
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
...