Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
Brain ; 131(Pt 3): 651-64, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18292081

RESUMEN

Non-steroidal anti-inflammatory agents (NSAIDs) are associated with a marked reduction in the risk of developing Alzheimer's disease, a form of dementia characterized by the accumulation of amyloid plaques containing the amyloid-beta protein (Abeta). Studies of the effects of NSAIDs upon the inflammatory response surrounding amyloid plaques and upon the generation of Abeta from the amyloid precursor protein (APP) have led to two proposed mechanisms by which NSAIDs may protect against Alzheimer's disease: one, the selective lowering of Abeta42 by a subset of NSAIDs; and two, the reduction of inflammation. Although Alzheimer's disease is a disorder of brain and synaptic function, the effects of NSAIDs on Abeta-mediated suppression of synaptic plasticity and memory function have never been reported. We therefore investigated how three different NSAIDs, chosen for their distinct effects on Abeta42 production and the inhibition of the cyclooxygenase (COX) isoenzymes, COX-1 and COX-2, affect memory function and synaptic plasticity. By focusing upon brain and synapse function, we made novel observations about the effects of NSAIDs on Abeta-mediated neural processes. Here we report that the selective inhibition of COX-2, but not COX-1, acutely prevented the suppression of hippocampal long-term plasticity (LTP) by Abeta. The non-selective NSAIDs, ibuprofen and naproxen, and a selective COX-2 inhibitor, MF-tricyclic, each restored memory function in Tg2576 mice over-expressing APP, and also blocked Abeta-mediated inhibition of LTP. There was no advantage of ibuprofen, a selective Abeta42-lowering agent (SALA), over the non-SALAs, naproxen and MF-tricyclic. The beneficial effects on memory did not depend upon lowered levels of Abeta42 or the inflammatory cytokines, tumour necrosis factor alpha (TNF-alpha) and interleukin 1beta (IL-1beta). Intriguingly, improved memory function was inversely related to prostaglandin E2 (PGE2) levels. Conversely, exogenous PGE2 prevented the restorative effects of COX-2 inhibitors on LTP. The data indicate that the inhibition of COX-2 blocks Abeta-mediated suppression of LTP and memory function, and that this block occurs independently of reductions in Abeta42 or decreases in inflammation. The results lead us to propose a third possible mechanism by which NSAIDs may protect against Alzheimer's disease, involving the blockade of a COX-2-mediated PGE2 response at synapses.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Inhibidores de la Ciclooxigenasa 2/farmacología , Memoria/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Péptidos beta-Amiloides/farmacología , Animales , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Inhibidores de la Ciclooxigenasa/farmacología , Inhibidores de la Ciclooxigenasa/uso terapéutico , Dinoprostona/fisiología , Furanos/farmacología , Furanos/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ibuprofeno/farmacología , Ibuprofeno/uso terapéutico , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/prevención & control , Ratones , Ratones Endogámicos C57BL , Naproxeno/farmacología , Naproxeno/uso terapéutico , Fragmentos de Péptidos/farmacología , Ratas , Sinapsis/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
3.
J Neurochem ; 103(4): 1594-607, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17760871

RESUMEN

Extracellular-signal regulated kinase (ERK) signaling is critical for memory and tightly regulated by acute environmental stimuli. In Alzheimer disease transgenic models, active ERK is shown to first be increased, then later reduced, but whether these baseline changes reflect disruptions in ERK signaling is less clear. We investigated the influence of the familial Alzheimer's disease transgene APPsw and beta-amyloid peptide (Abeta) immunoneutralization on cannulation injury-associated (i.c.v. infusion) ERK activation. At both 12 and 22 months of age, the trauma-associated activation of ERK observed in Tg(-) mice was dramatically attenuated in Tg(+). In cortices of 22-month-old non-infused mice, a reduction in ERK activation was observed in Tg(+), relative to Tg(-) mice. Intracerebroventricular (i.c.v.) anti-Abeta infusion significantly increased phosphorylated ERK, its substrate cAMP-response element-binding protein (CREB) and a downstream target, the NMDA receptor subunit. We also demonstrated that Abeta oligomer decreased active ERK and subsequently active CREB in human neuroblastoma cells, which could be prevented by oligomer immunoneutralization. Abeta oligomers also inhibited active ERK and CREB in primary neurons, in addition to reducing the downstream post-synaptic protein NMDA receptor subunit. These effects were reversed by anti-oligomer. Our data strongly support the existence of an APPsw transgene-dependent and Abeta oligomer-mediated defect in regulation of ERK activation.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Péptidos beta-Amiloides/fisiología , Proteína de Unión a CREB/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Sistema de Señalización de MAP Quinasas/genética , Transgenes/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Animales , Proteína de Unión a CREB/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Activación Enzimática/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Humanos , Ratones , Ratones Transgénicos
4.
Nat Neurosci ; 9(2): 234-42, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16415866

RESUMEN

Defects in dendritic spines are common to several forms of cognitive deficits, including mental retardation and Alzheimer disease. Because mutation of p21-activated kinase (PAK) can lead to mental retardation and because PAK-cofilin signaling is critical in dendritic spine morphogenesis and actin dynamics, we hypothesized that the PAK pathway is involved in synaptic and cognitive deficits in Alzheimer disease. Here, we show that PAK and its activity are markedly reduced in Alzheimer disease and that this is accompanied by reduced and redistributed phosphoPAK, prominent cofilin pathology and downstream loss of the spine actin-regulatory protein drebrin, which cofilin removes from actin. We found that beta-amyloid (Abeta) was directly involved in PAK signaling deficits and drebrin loss in Abeta oligomer-treated hippocampal neurons and in the Appswe transgenic mouse model bearing a double mutation leading to higher Abeta production. In addition, pharmacological PAK inhibition in adult mice was sufficient to cause similar cofilin pathology, drebrin loss and memory impairment, consistent with a potential causal role of PAK defects in cognitive deficits in Alzheimer disease.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Péptidos beta-Amiloides/metabolismo , Trastornos del Conocimiento/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Animales , Células Cultivadas , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Espinas Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Neuropéptidos/metabolismo , Ratas , Quinasas p21 Activadas
5.
J Neurosci Res ; 83(3): 374-84, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16385556

RESUMEN

Although active and passive immunization against the beta-amyloid peptide (Abeta) of amyloid plaque-bearing transgenic mice markedly reduces amyloid plaque deposition and improves cognition, the mechanisms of neuroprotection and impact on toxic oligomer species are not understood. We demonstrate that compared to control IgG2b, passive immunization with intracerebroventricular (icv) anti-Abeta (1-15) antibody into the AD HuAPPsw (Tg2576) transgenic mouse model reduced specific oligomeric forms of Abeta, including the dodecamers that correlate with cognitive decline. Interestingly, the reduction of soluble Abeta oligomers, but not insoluble Abeta, significantly correlated with reduced tau phosphorylation by glycogen synthase kinase-3beta (GSK-3beta), a major tau kinase implicated previously in mediating Abeta toxicity. A conformationally-directed antibody against amyloid oligomers (larger than tetramer) also reduced Abeta oligomer-induced activation of GSK3beta and protected human neuronal SH-SY5Y cells from Abeta oligomer-induced neurotoxicity, supporting a role for Abeta oligomers in human tau kinase activation. These data suggest that antibodies that are highly specific for toxic oligomer subspecies may reduce toxicity via reduction of GSK-3beta, which could be an important strategy for Alzheimer's disease (AD) therapeutics.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Péptidos beta-Amiloides/metabolismo , Anticuerpos/farmacología , Reactivadores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Western Blotting/métodos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Activación Enzimática/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Glucógeno Sintasa Quinasa 3 beta , Humanos , Inmunohistoquímica/métodos , Técnicas In Vitro , Masculino , Ratones , Ratones Transgénicos , Neuroblastoma , Fragmentos de Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Placa Amiloide/patología , Distribución Aleatoria , Tinción con Nitrato de Plata/métodos
6.
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
7.
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
8.
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
9.
Neuropsychopharmacology ; 30(6): 1111-20, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15688088

RESUMEN

Epidemiological and basic research suggests that nonsteroidal anti-inflammatory drugs (NSAIDs) should protect against the most common forms of Alzheimer's disease (AD). Ibuprofen reduces amyloid (Abeta) pathology in some transgenic models, but the precise mechanisms remain unclear. Although some reports show select NSAIDs inhibit amyloid production in vitro, the possibility that in vivo suppression of amyloid pathology occurs independent of Abeta production has not been ruled out. We show that ibuprofen reduced Abeta brain levels in rats from exogenously infused Abeta in the absence of altered Abeta production. To determine whether ibuprofen inhibits pro-amyloidogenic factors, APPsw (Tg2576) mice were treated with ibuprofen for 6 months, and expression levels of the Abeta and inflammation-related molecules alpha1 antichymotrypsin (ACT), apoE, BACE1, and peroxisome proliferator-activated receptor gamma) (PPARgamma) were measured. Among these, ACT, a factor whose overexpression accelerates amyloid pathology, was reduced by ibuprofen both in vivo and in vitro. IL-1beta, which was reduced in our animals by ibuprofen, induced mouse ACT in vitro. While some NSAIDs may inhibit Abeta42 production, these observations suggest that ibuprofen reduction of Abeta pathology may not be mediated by altered Abeta42 production. We present evidence supporting the hypothesis that ibuprofen-dependent amyloid reduction is mediated by inhibition of an alternate pathway (IL-1beta and its downstream target ACT).


Asunto(s)
Enfermedad de Alzheimer/patología , Antiinflamatorios no Esteroideos/farmacología , Ibuprofeno/farmacología , Interleucina-1/metabolismo , Placa Amiloide/patología , Inhibidores de Serina Proteinasa/farmacología , alfa 1-Antiquimotripsina/metabolismo , Proteínas de Fase Aguda/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Amiloidosis/patología , Amiloidosis/prevención & control , Animales , Apolipoproteínas E/biosíntesis , Ácido Aspártico Endopeptidasas , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitoma/metabolismo , Western Blotting , Endopeptidasas/biosíntesis , Inmunohistoquímica , Interleucina-1/farmacología , Masculino , Ratones , Ratones Transgénicos , Neuroglía/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , alfa 1-Antiquimotripsina/biosíntesis
10.
J Biol Chem ; 280(7): 5892-901, 2005 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-15590663

RESUMEN

Alzheimer's disease (AD) involves amyloid beta (Abeta) accumulation, oxidative damage, and inflammation, and risk is reduced with increased antioxidant and anti-inflammatory consumption. The phenolic yellow curry pigment curcumin has potent anti-inflammatory and antioxidant activities and can suppress oxidative damage, inflammation, cognitive deficits, and amyloid accumulation. Since the molecular structure of curcumin suggested potential Abeta binding, we investigated whether its efficacy in AD models could be explained by effects on Abeta aggregation. Under aggregating conditions in vitro, curcumin inhibited aggregation (IC(50) = 0.8 microM) as well as disaggregated fibrillar Abeta40 (IC(50) = 1 microM), indicating favorable stoichiometry for inhibition. Curcumin was a better Abeta40 aggregation inhibitor than ibuprofen and naproxen, and prevented Abeta42 oligomer formation and toxicity between 0.1 and 1.0 microM. Under EM, curcumin decreased dose dependently Abeta fibril formation beginning with 0.125 microM. The effects of curcumin did not depend on Abeta sequence but on fibril-related conformation. AD and Tg2576 mice brain sections incubated with curcumin revealed preferential labeling of amyloid plaques. In vivo studies showed that curcumin injected peripherally into aged Tg mice crossed the blood-brain barrier and bound plaques. When fed to aged Tg2576 mice with advanced amyloid accumulation, curcumin labeled plaques and reduced amyloid levels and plaque burden. Hence, curcumin directly binds small beta-amyloid species to block aggregation and fibril formation in vitro and in vivo. These data suggest that low dose curcumin effectively disaggregates Abeta as well as prevents fibril and oligomer formation, supporting the rationale for curcumin use in clinical trials preventing or treating AD.


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Curcumina/metabolismo , Curcumina/farmacología , Placa Amiloide/metabolismo , Placa Amiloide/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/toxicidad , Animales , Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Benzoatos/química , Benzoatos/farmacología , Benzotiazoles , Compuestos de Bifenilo/química , Compuestos de Bifenilo/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Rojo Congo , Curcumina/química , Diaminas/química , Diaminas/farmacología , Humanos , Ibuprofeno/farmacología , Ratones , Ratones Transgénicos , Naproxeno/farmacología , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Placa Amiloide/química , Placa Amiloide/genética , Unión Proteica/efectos de los fármacos , Desnaturalización Proteica/efectos de los fármacos , Estructura Cuaternaria de Proteína/efectos de los fármacos , Piridazinas/química , Piridazinas/farmacología , Solubilidad , Tiazoles
11.
J Neurosci ; 24(49): 11120-6, 2004 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-15590928

RESUMEN

Insulin-degrading enzyme (IDE) is one of the proteins that has been demonstrated to play a key role in degrading beta-amyloid (Abeta) monomer in vitro and in vivo, raising the possibility of upregulating IDE as an approach to reduce Abeta. Little is known, however, about the cellular and molecular regulation of IDE protein. Because one of the main functions of IDE is to degrade insulin, we hypothesized that there is a negative feedback mechanism whereby stimulation of insulin receptor-mediated signaling upregulates IDE to prevent chronic activation of the pathway. We show that treatment of primary hippocampal neurons with insulin increased IDE protein levels by approximately 25%. Insulin treatment also led to phosphatidylinositol-3 (PI3) kinase activation evidenced by Akt phosphorylation, which was blocked by PI3 kinase inhibitors, wortmannin and LY 294002. Inhibition of PI3 kinase abolished the IDE upregulation by insulin, indicating a cause-effect relationship between insulin signaling and IDE upregulation. Further support for this link was provided by the findings that deficient insulin signaling (decreased PI3 kinase subunit P85) was correlated with reduced IDE in Alzheimer's disease (AD) brains and in Tg2576 Swedish amyloid precursor protein transgenic mice fed a safflower oil-enriched ("Bad") diet used to accelerate pathogenesis. Consistent with IDE function in the degradation of Abeta monomer, the IDE decrease in the Bad diet-fed Tg2576 mice was associated with increased Abeta monomer levels. These in vitro and in vivo analyses validate the use of enhanced CNS insulin signaling as a potential strategy for AD intervention to correct the IDE defects occurring in AD.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Encéfalo/enzimología , Insulina/fisiología , Insulisina/metabolismo , Anciano , Anciano de 80 o más Años , Péptidos beta-Amiloides/metabolismo , Animales , Células Cultivadas , Diabetes Mellitus/metabolismo , Femenino , Hipocampo/enzimología , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Neuronas/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Insulina/fisiología , Aceite de Cártamo , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
12.
Neuron ; 43(5): 633-45, 2004 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-15339646

RESUMEN

Learning and memory depend on dendritic spine actin assembly and docosahexaenoic acid (DHA), an essential n-3 (omega-3) polyunsaturated fatty acid (PFA). High DHA consumption is associated with reduced Alzheimer's disease (AD) risk, yet mechanisms and therapeutic potential remain elusive. Here, we report that reduction of dietary n-3 PFA in an AD mouse model resulted in 80%-90% losses of the p85alpha subunit of phosphatidylinositol 3-kinase and the postsynaptic actin-regulating protein drebrin, as in AD brain. The loss of postsynaptic proteins was associated with increased oxidation, without concomitant neuron or presynaptic protein loss. n-3 PFA depletion increased caspase-cleaved actin, which was localized in dendrites ultrastructurally. Treatment of n-3 PFA-restricted mice with DHA protected against these effects and behavioral deficits and increased antiapoptotic BAD phosphorylation. Since n-3 PFAs are essential for p85-mediated CNS insulin signaling and selective protection of postsynaptic proteins, these findings have implications for neurodegenerative diseases where synaptic loss is critical, especially AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/prevención & control , Encéfalo/metabolismo , Dendritas/efectos de los fármacos , Dendritas/metabolismo , Ácidos Docosahexaenoicos/farmacología , Actinas/metabolismo , Enfermedad de Alzheimer/patología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Encéfalo/efectos de los fármacos , Encéfalo/patología , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/metabolismo , Dendritas/patología , Grasas Insaturadas en la Dieta/metabolismo , Grasas Insaturadas en la Dieta/farmacología , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/metabolismo , Ácidos Docosahexaenoicos/uso terapéutico , Regulación hacia Abajo/genética , Femenino , Alimentos Formulados , Humanos , Masculino , Ratones , Ratones Transgénicos , Microscopía Electrónica , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Subunidades de Proteína/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/patología , Proteína Letal Asociada a bcl
13.
J Neurosci Res ; 77(2): 217-28, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15211588

RESUMEN

There is increasing evidence that soluble amyloid-beta peptide (Abeta) uptake into neurons is an early event in the pathogenesis of Alzheimer's disease (AD). Identification of the early events leading to neuronal dysfunction is key to developing therapeutic strategies, but relative roles of receptors and factors modulating uptake are poorly understood. Studies have shown that transforming growth factor beta (TGFbeta), particularly TGFbeta2, can influence the targeting of Abeta to cells in vitro. TGFbeta2 can target Abeta to neurons in organotypic hippocampal slice cultures (OHSC). We examine a specific mechanism for TGFbeta2-mediated targeting of Abeta to neurons. The receptor-associated protein (RAP), a low-density lipoprotein receptor-related protein (LRP) antagonist, can attenuate the cellular targeting of Abeta both in vitro and in vivo and prevent Abeta/TGFbeta2-induced memory retention deficits. Using both in vitro and in vivo methods, we identify LRP as playing a role in TGFbeta2-mediated Abeta uptake, neurodegeneration, and spatial memory impairment.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Trastornos de la Memoria/metabolismo , Neuronas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Hipocampo/fisiopatología , Técnicas In Vitro , Proteína Asociada a Proteínas Relacionadas con Receptor de LDL/metabolismo , Proteína Asociada a Proteínas Relacionadas con Receptor de LDL/farmacología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/antagonistas & inhibidores , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Neuronas/patología , Transporte de Proteínas/fisiología , Factor de Crecimiento Transformador beta2
14.
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