Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Neurobiol Dis ; 82: 552-560, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26369878

RESUMEN

Increased dietary consumption of docosahexaenoic acid (DHA) is associated with decreased risk for Alzheimer's disease (AD). These effects have been postulated to arise from DHA's pleiotropic effects on AD pathophysiology, including its effects on ß-amyloid (Aß) production, aggregation, and toxicity. While in vitro studies suggest that DHA may inhibit and reverse the formation of toxic Aß oligomers, it remains uncertain whether these mechanisms operate in vivo at the physiological concentrations of DHA attainable through dietary supplementation. We sought to clarify the effects of dietary DHA supplementation on Aß indices in a transgenic APP/PS1 rat model of AD. Animals maintained on a DHA-supplemented diet exhibited reductions in hippocampal Aß plaque density and modest improvements on behavioral testing relative to those maintained on a DHA-depleted diet. However, DHA supplementation also increased overall soluble Aß oligomer levels in the hippocampus. Further quantification of specific conformational populations of Aß oligomers indicated that DHA supplementation increased fibrillar (i.e. putatively less toxic) Aß oligomers and decreased prefibrillar (i.e. putatively more toxic) Aß oligomers. These results provide in vivo evidence suggesting that DHA can modulate Aß aggregation by stabilizing soluble fibrillar Aß oligomers and thus reduce the formation of both Aß plaques and prefibrillar Aß oligomers. However, since fibrillar Aß oligomers still retain inherent neurotoxicity, DHA may need to be combined with other interventions that can additionally reduce fibrillar Aß oligomer levels for more effective prevention of AD in clinical settings.


Asunto(s)
Enfermedad de Alzheimer/dietoterapia , Péptidos beta-Amiloides/metabolismo , Suplementos Dietéticos , Ácidos Docosahexaenoicos , Hipocampo/metabolismo , Fragmentos de Péptidos/metabolismo , Placa Amiloide/dietoterapia , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Hipocampo/patología , Humanos , Masculino , Aprendizaje por Laberinto , Placa Amiloide/metabolismo , Placa Amiloide/patología , Placa Amiloide/psicología , Presenilina-1/genética , Presenilina-1/metabolismo , Multimerización de Proteína , Ratas Sprague-Dawley , Ratas Transgénicas , Resultado del Tratamiento
2.
J Neurosci ; 34(21): 7124-36, 2014 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-24849348

RESUMEN

Hyperphosphorylation and accumulation of tau aggregates are prominent features in tauopathies, including Alzheimer's disease, but the impact of loss of tau function on synaptic and cognitive deficits remains poorly understood. We report that old (19-20 months; OKO) but not middle-aged (8-9 months; MKO) tau knock-out mice develop Morris Water Maze (MWM) deficits and loss of hippocampal acetylated α-tubulin and excitatory synaptic proteins. Mild motor deficits and reduction in tyrosine hydroxylase (TH) in the substantia nigra were present by middle age, but did not affect MWM performance, whereas OKO mice showed MWM deficits paralleling hippocampal deficits. Deletion of tau, a microtubule-associated protein (MAP), resulted in increased levels of MAP1A, MAP1B, and MAP2 in MKO, followed by loss of MAP2 and MAP1B in OKO. Hippocampal synaptic deficits in OKO mice were partially corrected with dietary supplementation with docosahexaenoic acid (DHA) and both MWM and synaptic deficits were fully corrected by combining DHA with α-lipoic acid (ALA), which also prevented TH loss. DHA or DHA/ALA restored phosphorylated and total GSK3ß and attenuated hyperactivation of the tau C-Jun N-terminal kinases (JNKs) while increasing MAP1B, dephosphorylated (active) MAP2, and acetylated α-tubulin, suggesting improved microtubule stability and maintenance of active compensatory MAPs. Our results implicate the loss of MAP function in age-associated hippocampal deficits and identify a safe dietary intervention, rescuing both MAP function and TH in OKO mice. Therefore, in addition to microtubule-stabilizing therapeutic drugs, preserving or restoring compensatory MAP function may be a useful new prevention strategy.


Asunto(s)
Envejecimiento/patología , Hipocampo/patología , Aprendizaje por Laberinto/fisiología , Sinapsis/metabolismo , Proteínas tau/deficiencia , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hipocampo/efectos de los fármacos , Discapacidades para el Aprendizaje/dietoterapia , Discapacidades para el Aprendizaje/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos del Movimiento/dietoterapia , Trastornos del Movimiento/etiología , Desempeño Psicomotor/fisiología , Tiempo de Reacción/efectos de los fármacos , Sustancia Negra/metabolismo , Sustancia Negra/patología , Sinapsis/efectos de los fármacos , Sinapsis/genética , Ácido Tióctico/administración & dosificación
3.
J Neurosci ; 29(28): 9078-89, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19605645

RESUMEN

Both insulin resistance (type II diabetes) and beta-amyloid (Abeta) oligomers are implicated in Alzheimer's disease (AD). Here, we investigate the role of Abeta oligomer-induced c-Jun N-terminal kinase (JNK) activation leading to phosphorylation and degradation of the adaptor protein insulin receptor substrate-1 (IRS-1). IRS-1 couples insulin and other trophic factor receptors to downstream kinases and neuroprotective signaling. Increased phospho-IRS-1 is found in AD brain and insulin-resistant tissues from diabetics. Here, we report Abeta oligomers significantly increased active JNK and phosphorylation of IRS-1 (Ser616) and tau (Ser422) in cultured hippocampal neurons, whereas JNK inhibition blocked these responses. The omega-3 fatty acid docosahexaenoic acid (DHA) similarly inhibited JNK and the phosphorylation of IRS-1 and tau in cultured hippocampal neurons. Feeding 3xTg-AD transgenic mice a diet high in saturated and omega-6 fat increased active JNK and phosphorylated IRS-1 and tau. Treatment of the 3xTg-AD mice on high-fat diet with fish oil or curcumin or a combination of both for 4 months reduced phosphorylated JNK, IRS-1, and tau and prevented the degradation of total IRS-1. This was accompanied by improvement in Y-maze performance. Mice fed with fish oil and curcumin for 1 month had more significant effects on Y-maze, and the combination showed more significant inhibition of JNK, IRS-1, and tau phosphorylation. These data indicate JNK mediates Abeta oligomer inactivation of IRS-1 and phospho-tau pathology and that dietary treatment with fish oil/DHA, curcumin, or a combination of both has the potential to improve insulin/trophic signaling and cognitive deficits in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/farmacología , Curcumina/farmacología , Inhibidores Enzimáticos/farmacología , Ácidos Grasos Omega-3/farmacología , Proteínas Sustrato del Receptor de Insulina/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/dietoterapia , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Conducta Animal , Células Cultivadas , Curcumina/uso terapéutico , Modelos Animales de Enfermedad , Embrión de Mamíferos , Inhibidores Enzimáticos/uso terapéutico , Ácidos Grasos Omega-3/uso terapéutico , Hipocampo/citología , Humanos , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Fosforilación/efectos de los fármacos , Cambios Post Mortem , Presenilina-1/genética , Ratas , Ratas Sprague-Dawley , Serina/metabolismo
4.
J Pharmacol Exp Ther ; 326(1): 196-208, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18417733

RESUMEN

Curcumin can reduce inflammation and neurodegeneration, but its chemical instability and metabolism raise concerns, including whether the more stable metabolite tetrahydrocurcumin (TC) may mediate efficacy. We examined the antioxidant, anti-inflammatory, or anti-amyloidogenic effects of dietary curcumin and TC, either administered chronically to aged Tg2576 APPsw mice or acutely to lipopolysaccharide (LPS)-injected wild-type mice. Despite dramatically higher drug plasma levels after TC compared with curcumin gavage, resulting brain levels of parent compounds were similar, correlating with reduction in LPS-stimulated inducible nitric-oxide synthase, nitrotyrosine, F2 isoprostanes, and carbonyls. In both the acute (LPS) and chronic inflammation (Tg2576), TC and curcumin similarly reduced interleukin-1beta. Despite these similarities, only curcumin was effective in reducing amyloid plaque burden, insoluble beta-amyloid peptide (Abeta), and carbonyls. TC had no impact on plaques or insoluble Abeta, but both reduced Tris-buffered saline-soluble Abeta and phospho-c-Jun NH(2)-terminal kinase (JNK). Curcumin but not TC prevented Abeta aggregation. The TC metabolite was detected in brain and plasma from mice chronically fed the parent compound. These data indicate that the dienone bridge present in curcumin, but not in TC, is necessary to reduce plaque deposition and protein oxidation in an Alzheimer's model. Nevertheless, TC did reduce neuroinflammation and soluble Abeta, effects that may be attributable to limiting JNK-mediated transcription. Because of its favorable safety profile and the involvement of misfolded proteins, oxidative damage, and inflammation in multiple chronic degenerative diseases, these data relating curcumin dosing to the blood and tissue levels required for efficacy should help translation efforts from multiple successful preclinical models.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Curcumina/metabolismo , Curcumina/uso terapéutico , Modelos Animales de Enfermedad , Enfermedad de Alzheimer/patología , Animales , Disponibilidad Biológica , Curcumina/química , Femenino , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Relación Estructura-Actividad
5.
Neurobiol Aging ; 26 Suppl 1: 133-6, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16266772

RESUMEN

Alzheimer's disease (AD) and cardiovascular disease (CVD) are syndromes of aging that share analogous lesions and risk factors, involving lipoproteins, oxidative damage and inflammation. Unlike in CVD, in AD, sensitive biomarkers are unknown, and high-risk groups are understudied. To identify potential prevention strategies in AD, we have focused on pre-clinical models (transgenic and amyloid infusion models), testing dietary/lifestyle factors strongly implicated in reducing risk in epidemiological studies. Initially, we reported the impact of non-steroidal anti-inflammatory drugs (NSAIDs), notably ibuprofen, which reduced amyloid accumulation, but suppressed few inflammatory markers and without reducing oxidative damage. Safety concerns with chronic NSAIDs led to a screen of alternative NSAIDs and identification of the phenolic anti-inflammatory/anti-oxidant compound curcumin, the yellow pigment in turmeric that we found targeted multiple AD pathogenic cascades. The dietary omega-3 fatty acid, docosahexaenoic acid (DHA), also limited amyloid, oxidative damage and synaptic and cognitive deficits in a transgenic mouse model. Both DHA and curcumin have favorable safety profiles, epidemiology and efficacy, and may exert general anti-aging benefits (anti-cancer and cardioprotective.).


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Antioxidantes/uso terapéutico , Ácidos Grasos Omega-3/uso terapéutico , Flavonoides/uso terapéutico , Fenoles/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Curcumina/uso terapéutico , Humanos , Polifenoles
6.
Eur J Neurosci ; 22(3): 617-26, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16101743

RESUMEN

Epidemiological data indicate that low n-3 polyunsaturated fatty acids (PFA) intake is a readily manipulated dietary risk factor for Alzheimer's disease (AD). Studies in animals confirm the deleterious effect of n-3 PFA depletion on cognition and on dendritic scaffold proteins. Here, we show that in transgenic mice overexpressing the human AD gene APPswe (Tg2576), safflower oil-induced n-3 PFA deficiency caused a decrease in N-methyl-D-aspartate (NMDA) receptor subunits, NR2A and NR2B, in the cortex and hippocampus with no loss of the presynaptic markers, synaptophysin and synaptosomal-associated protein 25 (SNAP-25). n-3 PFA depletion also decreased the NR1 subunit in the hippocampus and Ca2+/calmodulin-dependent protein kinase (CaMKII) in the cortex of Tg2576 mice. These effects of dietary n-3 PFA deficiency were greatly amplified in Tg2576 mice compared to nontransgenic mice. Loss of the NR2B receptor subunit was not explained by changes in mRNA expression, but correlated with p85alpha phosphatidylinositol 3-kinase levels. Most interestingly, n-3 PFA deficiency dramatically increased levels of protein fragments, corresponding to caspase/calpain-cleaved fodrin and gelsolin in Tg2576 mice. This effect was minimal in nontransgenic mice suggesting that n-3 PFA depletion potentiated caspase activation in the Tg2576 mouse model of AD. Dietary supplementation with docosahexaenoic acid (DHA; 22 : 6n-3) partly protected from NMDA receptor subunit loss and accumulation of fodrin and gelsolin fragments but fully prevented CaMKII decrease. The marked effect of dietary n-3 PFA on NMDA receptors and caspase/calpain activation in the cortex of an animal model of AD provide new insights into how dietary essential fatty acids may influence cognition and AD risk.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Caspasas/metabolismo , Ácidos Grasos Insaturados/deficiencia , Receptores de N-Metil-D-Aspartato/metabolismo , Triglicéridos/deficiencia , Alcaloides/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Western Blotting/métodos , Encéfalo/efectos de los fármacos , Química Encefálica/efectos de los fármacos , Química Encefálica/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Calpaína/metabolismo , Proteínas Portadoras/metabolismo , Dieta Reductora/métodos , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/farmacología , Ácidos Grasos/análisis , Ácidos Grasos Omega-3 , Femenino , Gelsolina/metabolismo , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , ARN Mensajero/metabolismo , Receptores de N-Metil-D-Aspartato/clasificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Estadística como Asunto , Proteína 25 Asociada a Sinaptosomas , Factores de Tiempo , Proteína Letal Asociada a bcl
7.
J Neurosci ; 25(12): 3032-40, 2005 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-15788759

RESUMEN

Epidemiological studies suggest that increased intake of the omega-3 (n-3) polyunsaturated fatty acid (PUFA) docosahexaenoic acid (DHA) is associated with reduced risk of Alzheimer's disease (AD). DHA levels are lower in serum and brains of AD patients, which could result from low dietary intake and/or PUFA oxidation. Because effects of DHA on Alzheimer pathogenesis, particularly on amyloidosis, are unknown, we used the APPsw (Tg2576) transgenic mouse model to evaluate the impact of dietary DHA on amyloid precursor protein (APP) processing and amyloid burden. Aged animals (17-19 months old) were placed in one of three groups until 22.5 months of age: control (0.09% DHA), low-DHA (0%), or high-DHA (0.6%) chow. beta-Amyloid (Abeta) ELISA of the detergent-insoluble extract of cortical homogenates showed that DHA-enriched diets significantly reduced total Abeta by >70% when compared with low-DHA or control chow diets. Dietary DHA also decreased Abeta42 levels below those seen with control chow. Image analysis of brain sections with an antibody against Abeta (amino acids 1-13) revealed that overall plaque burden was significantly reduced by 40.3%, with the largest reductions (40-50%) in the hippocampus and parietal cortex. DHA modulated APP processing by decreasing both alpha- and beta-APP C-terminal fragment products and full-length APP. BACE1 (beta-secretase activity of the beta-site APP-cleaving enzyme), ApoE (apolipoprotein E), and transthyretin gene expression were unchanged with the high-DHA diet. Together, these results suggest that dietary DHA could be protective against beta-amyloid production, accumulation, and potential downstream toxicity.


Asunto(s)
Enfermedad de Alzheimer/dietoterapia , Enfermedad de Alzheimer/metabolismo , Amiloide/metabolismo , Ácidos Docosahexaenoicos/administración & dosificación , Administración Oral , Secretasas de la Proteína Precursora del Amiloide , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Ácido Aspártico Endopeptidasas , Western Blotting/métodos , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Diagnóstico por Imagen/métodos , Grasas Insaturadas en la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/farmacología , Relación Dosis-Respuesta a Droga , Endopeptidasas/genética , Endopeptidasas/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Ácidos Grasos/metabolismo , Inmunohistoquímica/métodos , Masculino , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/metabolismo , Placa Amiloide/patología , Prealbúmina/genética , Prealbúmina/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Estadística como Asunto
8.
Ann N Y Acad Sci ; 1035: 68-84, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15681801

RESUMEN

Both oxidative damage and inflammation are elevated in brains of Alzheimer's disease (AD) patients, but their pathogenic significance remains unclear. The reduced AD risk associated with high intake of both nonsteroidal anti-inflammatory drugs (NSAIDs) and antioxidants suggests causal roles, but clinical trials in AD patients have yielded only limited or negative results. To test the potential efficacy and mechanisms of candidate approaches, we have explored conventional and unconventional NSAIDs, antioxidants, and combined NSAID/antioxidants in cell culture and animal models for AD (including aging APPsw transgenic mice and soluble Abeta rodent infusion models). The conventional NSAID ibuprofen has the strongest epidemiological support. At sustainable doses designed to mimic protective consumption in the epidemiology, ibuprofen reduces amyloid accumulation but suppresses a surprisingly limited subset of inflammatory markers in APPsw transgenic mice. Both Ab production (APP, beta- and gamma-secretases) and post-production pathways (those affecting Abeta aggregation or clearance: e.g., IL-1 or alpha1ACT) are potentially involved in ibuprofen and other NSAID anti-AD activities. The post-production pathways are predictably shared with other seemingly protective NSAIDs, including naproxen that do not lower Abeta42 in vitro. Using clinically feasible dosing, brain levels of NSAIDs appear too low to implicate a number of pharmacological dose targets that have been demonstrated in vitro. Ibuprofen did not suppress microglial markers related to phagocytosis. The putative anti-inflammatory omega-3 fatty acid DHA had a profound impact on pathogenesis but did not lower inflammation, while vitamin E was surprisingly ineffective in reducing oxidative damage or amyloid in the aged APPsw mouse. In contrast, the unconventional NSAID/antioxidant curcumin was effective, lowering oxidative damage, cognitive deficits, synaptic marker loss, and amyloid deposition. Curcumin proved to be immunomodulatory, simultaneously inhibiting cytokine and microglial activation indices related to neurotoxicity, but increasing an index of phagocytosis. Curcumin directly targeted Abeta and was also effective in other models, warranting further preclinical and clinical exploration.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Antiinflamatorios no Esteroideos/uso terapéutico , Antioxidantes/uso terapéutico , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Ácidos Grasos Omega-3/uso terapéutico , Humanos , Técnicas In Vitro , Inflamación/tratamiento farmacológico , Modelos Biológicos , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA