Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Neurodegener ; 13(1): 22, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29764453

RESUMO

BACKGROUND: Extracellular aggregation of the amyloid-ß (Aß) peptide into toxic multimers is a key event in Alzheimer's disease (AD) pathogenesis. Aß aggregation is concentration-dependent, with higher concentrations of Aß much more likely to form toxic species. The processes that regulate extracellular levels of Aß therefore stand to directly affect AD pathology onset. Studies from our lab and others have demonstrated that synaptic activity is a critical regulator of Aß production through both presynaptic and postsynaptic mechanisms. AMPA receptors (AMPA-Rs), as the most abundant ionotropic glutamate receptors, have the potential to greatly impact Aß levels. METHODS: In order to study the role of AMPA-Rs in Aß regulation, we used in vivo microdialysis in an APP/PS1 mouse model to simultaneously deliver AMPA and other treatments while collecting Aß from the interstitial fluid (ISF). Changes in Aß production and clearance along with inflammation were assessed using biochemical approaches. IL-6 deficient mice were utilized to test the role of IL-6 signaling in AMPA-R-mediated regulation of Aß levels. RESULTS: We found that AMPA-R activation decreases in ISF Aß levels in a dose-dependent manner. Moreover, the effect of AMPA treatment involves three distinct pathways. Steady-state activity of AMPA-Rs normally promotes higher ISF Aß. Evoked AMPA-R activity, however, decreases Aß levels by both stimulating glutamatergic transmission and activating downstream NMDA receptor (NMDA-R) signaling and, with extended AMPA treatment, acting independently of NMDA-Rs. Surprisingly, we found this latter, direct AMPA pathway of Aß regulation increases Aß clearance, while Aß production appears to be largely unaffected. Furthermore, the AMPA-dependent decrease is not observed in IL-6 deficient mice, indicating a role for IL-6 signaling in AMPA-R-mediated Aß clearance. CONCLUSION: Though basal levels of AMPA-R activity promote higher levels of ISF Aß, evoked AMPA-R signaling decreases Aß through both NMDA-R-dependent and -independent pathways. We find that evoked AMPA-R signaling increases clearance of extracellular Aß, at least in part through enhanced IL-6 signaling. These data emphasize that Aß regulation by synaptic activity involves a number of independent pathways that together determine extracellular Aß levels. Understanding how these pathways maintain Aß levels prior to AD pathology may provide insights into disease pathogenesis.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Receptores de AMPA/metabolismo , Animais , Modelos Animais de Doenças , Interleucina-6/metabolismo , Camundongos , Camundongos Transgênicos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/metabolismo
2.
J Neurosci ; 37(15): 4023-4031, 2017 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-28275161

RESUMO

Accumulation and deposition of amyloid-ß (Aß) in the brain represent an early and perhaps necessary step in the pathogenesis of Alzheimer's disease (AD). Aß accumulation leads to the formation of Aß aggregates, which may directly and indirectly lead to eventual neurodegeneration. While Aß production is accelerated in many familial forms of early-onset AD, increasing evidence indicates that impaired clearance of Aß is more evident in late-onset AD. To uncover the mechanisms underlying impaired Aß clearance in AD, we examined the role of low-density lipoprotein receptor-related protein 1 (LRP1) in astrocytes. Although LRP1 has been shown to play critical roles in brain Aß metabolism in neurons and vascular mural cells, its role in astrocytes, the most abundant cell type in the brain responsible for maintaining neuronal homeostasis, remains unclear. Here, we show that astrocytic LRP1 plays a critical role in brain Aß clearance. LRP1 knockdown in primary astrocytes resulted in decreased cellular Aß uptake and degradation. In addition, silencing of LRP1 in astrocytes led to downregulation of several major Aß-degrading enzymes, including matrix metalloproteases MMP2, MMP9, and insulin-degrading enzyme. More important, conditional knock-out of the Lrp1 gene in astrocytes in the background of APP/PS1 mice impaired brain Aß clearance, exacerbated Aß accumulation, and accelerated amyloid plaque deposition without affecting its production. Together, our results demonstrate that astrocytic LRP1 plays an important role in Aß metabolism and that restoring LRP1 expression and function in the brain could be an effective strategy to facilitate Aß clearance and counter amyloid pathology in AD.SIGNIFICANCE STATEMENT Astrocytes represent a major cell type regulating brain homeostasis; however, their roles in brain clearance of amyloid-ß (Aß) and underlying mechanism are not clear. In this study, we used both cellular models and conditional knock-out mouse models to address the role of a critical Aß receptor, the low-density lipoprotein receptor-related protein 1 (LRP1) in astrocytes. We found that LRP1 in astrocytes plays a critical role in brain Aß clearance by modulating several Aß-degrading enzymes and cellular degradation pathways. Our results establish a critical role of astrocytic LRP1 in brain Aß clearance and shed light on specific Aß clearance pathways that may help to establish new targets for AD prevention and therapy.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Encéfalo/metabolismo , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/metabolismo , Receptores de LDL/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Astrócitos/patologia , Encéfalo/patologia , Células Cultivadas , Feminino , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Masculino , Camundongos , Camundongos Knockout
3.
Cell Chem Biol ; 23(12): 1526-1538, 2016 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-27889409

RESUMO

Pharmacological screening in physiologically relevant brain cells is crucial for identifying neuroactive compounds that better translate into in vivo biology and efficacious therapeutics. Pharmacological enhancement of apolipoprotein E (apoE), a cholesterol-transporting apolipoprotein, has been proposed as a promising therapeutic approach for Alzheimer's disease. Several nuclear receptor agonists were initially shown to increase brain apoE levels together with ATP-binding cassette transporter 1 (ABCA1), but their underlying mechanisms remain unclear. To gain an insight on brain apoE regulation, we performed an unbiased high-throughput screening of known drugs and bioactive compounds in cultured human primary astrocytes, the major apoE-producing cell type in the brain. We have identified several small molecules that increase apoE secretion via previously unknown mechanisms, including those not co-inducing ABCA1. These newly identified compounds are active preferentially in human astrocytes but not in an astrocytoma cell line, furnishing new tools for investigating biological pathways underlying brain apoE production.

4.
Neurobiol Dis ; 85: 218-224, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26563933

RESUMO

Physical activity has long been hypothesized to influence the risk and pathology of Alzheimer's disease. However, the amount of physical activity necessary for these benefits is unclear. We examined the effects of three months of low and high intensity exercise training on soluble Aß40 and Aß42 levels in extracellular enriched fractions from the cortex and hippocampus of young Tg2576 mice. Low (LOW) and high (HI) intensity exercise training animals ran at speeds of 15m/min on a level treadmill and 32 m/min at a 10% grade, respectively for 60 min per day, five days per week, from three to six months of age. Sedentary mice (SED) were placed on a level, non-moving, treadmill for the same duration. Soleus muscle citrate synthase activity increased by 39% in the LOW group relative to SED, and by 71% in the HI group relative to LOW, indicating an exercise training effect in these mice. Soluble Aß40 concentrations decreased significantly in an exercise training dose-dependent manner in the cortex. In the hippocampus, concentrations were decreased significantly in the HI group relative to LOW and SED. Soluble Aß42 levels also decreased significantly in an exercise training dose-dependent manner in both the cortex and hippocampus. Five proteins involved in Aß clearance (neprilysin, IDE, MMP9, LRP1 and HSP70) were elevated by exercise training with its intensity playing a role in each case. Our data demonstrate that exercise training reduces extracellular soluble Aß in the brains of Tg2576 mice in a dose-dependent manner through an up-regulation of Aß clearance.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Terapia por Exercício/métodos , Atividade Motora , Fragmentos de Peptídeos/metabolismo , Animais , Córtex Cerebral/metabolismo , Citrato (si)-Sintase/metabolismo , Modelos Animais de Doenças , Proteínas de Choque Térmico HSP70/metabolismo , Hipocampo/efeitos dos fármacos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Neprilisina/metabolismo , RNA Mensageiro/metabolismo , Distribuição Aleatória , Receptores de LDL/metabolismo , Resultado do Tratamento , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima
5.
J Neurosci ; 35(35): 12137-51, 2015 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-26338325

RESUMO

In AD, an imbalance between Aß production and removal drives elevated brain Aß levels and eventual amyloid plaque deposition. APP undergoes nonamyloidogenic processing via α-cleavage at the plasma membrane, amyloidogenic ß- and γ-cleavage within endosomes to generate Aß, or lysosomal degradation in neurons. Considering multiple reports implicating impaired lysosome function as a driver of increased amyloidogenic processing of APP, we explored the efficacy of targeting transcription factor EB (TFEB), a master regulator of lysosomal pathways, to reduce Aß levels. CMV promoter-driven TFEB, transduced via stereotactic hippocampal injections of adeno-associated virus particles in APP/PS1 mice, localized primarily to neuronal nuclei and upregulated lysosome biogenesis. This resulted in reduction of APP protein, the α and ß C-terminal APP fragments (CTFs), and in the steady-state Aß levels in the brain interstitial fluid. In aged mice, total Aß levels and amyloid plaque load were selectively reduced in the TFEB-transduced hippocampi. TFEB transfection in N2a cells stably expressing APP695, stimulated lysosome biogenesis, reduced steady-state levels of APP and α- and ß-CTFs, and attenuated Aß generation by accelerating flux through the endosome-lysosome pathway. Cycloheximide chase assays revealed a shortening of APP half-life with exogenous TFEB expression, which was prevented by concomitant inhibition of lysosomal acidification. These data indicate that TFEB enhances flux through lysosomal degradative pathways to induce APP degradation and reduce Aß generation. Activation of TFEB in neurons is an effective strategy to attenuate Aß generation and attenuate amyloid plaque deposition in AD. SIGNIFICANCE STATEMENT: A key driver for AD pathogenesis is the net balance between production and clearance of Aß, the major component of amyloid plaques. Here we demonstrate that lysosomal degradation of holo-APP influences Aß production by limiting the availability of APP for amyloidogenic processing. Using viral gene transfer of transcription factor EB (TFEB), a master regulator of lysosome biogenesis in neurons of APP/PS1 mice, steady-state levels of APP were reduced, resulting in decreased interstitial fluid Aß levels and attenuated amyloid deposits. These effects were caused by accelerated lysosomal degradation of endocytosed APP, reflected by reduced APP half-life and steady-state levels in TFEB-expressing cells, with resultant decrease in Aß production and release. Additional studies are needed to explore the therapeutic potential of this approach.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Lisossomos/metabolismo , Neurônios/metabolismo , Placa Amiloide/metabolismo , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Encéfalo/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular Tumoral , Dependovirus/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Lisossomos/genética , Lisossomos/patologia , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Mutação/genética , Neuroblastoma/patologia , Neurônios/patologia , Placa Amiloide/genética , Placa Amiloide/patologia , Presenilina-1/genética
6.
J Am Heart Assoc ; 3(6): e001156, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25392541

RESUMO

BACKGROUND: Brain lipoprotein metabolism is dependent on lipoprotein particles that resemble plasma high-density lipoproteins but that contain apolipoprotein (apo) E rather than apoA-I as their primary protein component. Astrocytes and microglia secrete apoE but not apoA-I; however, apoA-I is detectable in both cerebrospinal fluid and brain tissue lysates. The route by which plasma apoA-I enters the central nervous system is unknown. METHODS AND RESULTS: Steady-state levels of murine apoA-I in cerebrospinal fluid and interstitial fluid are 0.664 and 0.120 µg/mL, respectively, whereas brain tissue apoA-I is ≈10% to 15% of its levels in liver. Recombinant, fluorescently tagged human apoA-I injected intravenously into mice localizes to the choroid plexus within 30 minutes and accumulates in a saturable, dose-dependent manner in the brain. Recombinant, fluorescently tagged human apoA-I accumulates in the brain for 2 hours, after which it is eliminated with a half-life of 10.3 hours. In vitro, human apoA-I is specifically bound, internalized, and transported across confluent monolayers of primary human choroid plexus epithelial cells and brain microvascular endothelial cells. CONCLUSIONS: Following intravenous injection, recombinant human apoA-I rapidly localizes predominantly to the choroid plexus. Because apoA-I mRNA is undetectable in murine brain, our results suggest that plasma apoA-I, which is secreted from the liver and intestine, gains access to the central nervous system primarily by crossing the blood-cerebrospinal fluid barrier via specific cellular mediated transport, although transport across the blood-brain barrier may also contribute to a lesser extent.


Assuntos
Apolipoproteína A-I/administração & dosagem , Apolipoproteína A-I/farmacocinética , Barreira Hematoencefálica/metabolismo , Plexo Corióideo/metabolismo , Animais , Apolipoproteína A-I/sangue , Apolipoproteína A-I/líquido cefalorraquidiano , Apolipoproteína A-I/genética , Transporte Biológico , Permeabilidade Capilar , Células Cultivadas , Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Feminino , Meia-Vida , Humanos , Injeções Intravenosas , Taxa de Depuração Metabólica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacocinética , Distribuição Tecidual
7.
J Neurosci ; 34(29): 9607-20, 2014 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-25031402

RESUMO

In sporadic Alzheimer's disease (AD), impaired Aß removal contributes to elevated extracellular Aß levels that drive amyloid plaque pathogenesis. Extracellular proteolysis, export across the blood-brain barrier, and cellular uptake facilitate physiologic Aß clearance. Astrocytes can take up and degrade Aß, but it remains unclear whether this function is insufficient in AD or can be enhanced to accelerate Aß removal. Additionally, age-related dysfunction of lysosomes, the major degradative organelles wherein Aß localizes after uptake, has been implicated in amyloid plaque pathogenesis. We tested the hypothesis that enhancing lysosomal function in astrocytes with transcription factor EB (TFEB), a master regulator of lysosome biogenesis, would promote Aß uptake and catabolism and attenuate plaque pathogenesis. Exogenous TFEB localized to the nucleus with transcriptional induction of lysosomal biogenesis and function in vitro. This resulted in significantly accelerated uptake of exogenously applied Aß42, with increased localization to and degradation within lysosomes in C17.2 cells and primary astrocytes, indicating that TFEB is sufficient to coordinately enhance uptake, trafficking, and degradation of Aß. Stereotactic injection of adeno-associated viral particles carrying TFEB driven by a glial fibrillary acidic protein promoter was used to achieve astrocyte-specific expression in the hippocampus of APP/PS1 transgenic mice. Exogenous TFEB localized to astrocyte nuclei and enhanced lysosome function, resulting in reduced Aß levels and shortened half-life in the brain interstitial fluid and reduced amyloid plaque load in the hippocampus compared with control virus-injected mice. Therefore, activation of TFEB in astrocytes is an effective strategy to restore adequate Aß removal and counter amyloid plaque pathogenesis in AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Astrócitos/citologia , Astrócitos/metabolismo , Lisossomos/metabolismo , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/tratamento farmacológico , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Recém-Nascidos , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Córtex Cerebral/citologia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Placa Amiloide/genética , Placa Amiloide/metabolismo , Presenilina-1/genética , Transfecção
8.
J Alzheimers Dis ; 38(4): 767-86, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24072071

RESUMO

The formation and accumulation of toxic amyloid-ß peptides (Aß) in the brain may drive the pathogenesis of Alzheimer's disease. Accordingly, disease-modifying therapies for Alzheimer's disease and related disorders could result from treatments regulating Aß homeostasis. Examples are the inhibition of production, misfolding, and accumulation of Aß or the enhancement of its clearance. Here we show that oral treatment with ACI-91 (Pirenzepine) dose-dependently reduced brain Aß burden in AßPPPS1, hAßPPSL, and AßPP/PS1 transgenic mice. A possible mechanism of action of ACI-91 may occur through selective inhibition of muscarinic acetylcholine receptors (AChR) on endothelial cells of brain microvessels and enhanced Aß peptide clearance across the blood-brain barrier. One month treatment with ACI-91 increased the clearance of intrathecally-injected Aß in plaque-bearing mice. ACI-91 also accelerated the clearance of brain-injected Aß in blood and peripheral tissues by favoring its urinal excretion. A single oral dose of ACI-91 reduced the half-life of interstitial Aß peptide in pre-plaque mhAßPP/PS1d mice. By extending our studies to an in vitro model, we showed that muscarinic AChR inhibition by ACI-91 and Darifenacin augmented the capacity of differentiated endothelial monolayers for active transport of Aß peptide. Finally, ACI-91 was found to consistently affect, in vitro and in vivo, the expression of endothelial cell genes involved in Aß transport across the Blood Brain Brain (BBB). Thus increased Aß clearance through the BBB may contribute to reduced Aß burden and associated phenotypes. Inhibition of muscarinic AChR restricted to the periphery may present a therapeutic advantage as it avoids adverse central cholinergic effects.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/metabolismo , Angiopatia Amiloide Cerebral/metabolismo , Modelos Animais de Doenças , Antagonistas Muscarínicos/uso terapêutico , Fenótipo , Receptores Muscarínicos/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Angiopatia Amiloide Cerebral/tratamento farmacológico , Angiopatia Amiloide Cerebral/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Antagonistas Muscarínicos/farmacologia , Pirenzepina/farmacologia , Pirenzepina/uso terapêutico
9.
J Neurosci ; 33(49): 19276-83, 2013 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-24305823

RESUMO

Alzheimer's disease (AD) is the most prevalent form of dementia in the elderly population. Accumulation, aggregation, and deposition of amyloid-ß (Aß) peptides generated through proteolytic cleavage of amyloid precursor protein (APP) are likely initiating events in the pathogenesis of AD. While Aß production is accelerated in familial AD, increasing evidence indicates that impaired clearance of Aß is responsible for late-onset AD. Because Aß is mainly generated in neurons, these cells are predicted to have the highest risk of encountering Aß among all cell types in the brain. However, it is still unclear whether they are also involved in Aß clearance. Here we show that receptor-mediated endocytosis in neurons by the low-density lipoprotein receptor-related protein 1 (LRP1) plays a critical role in brain Aß clearance. LRP1 is known to be an endocytic receptor for multiple ligands including Aß. Conditional knock-out of Lrp1 in mouse forebrain neurons leads to increased brain Aß levels and exacerbated amyloid plaque deposition selectively in the cortex of amyloid model APP/PS1 mice without affecting Aß production. In vivo microdialysis studies demonstrated that Aß clearance in brain interstitial fluid is impaired in neuronal Lrp1 knock-out mice. Because the neuronal LRP1-deletion did not affect the mRNA levels of major Aß degrading enzymes, neprilysin and insulin-degrading enzyme, the disturbed Aß clearance is likely due to the suppression of LRP1-mediated neuronal Aß uptake and degradation. Together, our results demonstrate that LRP1 plays an important role in receptor-mediated clearance of Aß and indicate that neurons not only produce but also clear Aß.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Neurônios/metabolismo , Receptores de LDL/metabolismo , Vesículas Transportadoras/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Western Blotting , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Ensaio de Imunoadsorção Enzimática , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Camundongos Knockout , Microscopia Confocal , Reação em Cadeia da Polimerase , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Interferente Pequeno/genética
10.
J Biol Chem ; 287(25): 21279-89, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22539346

RESUMO

One of the pathological hallmarks of Alzheimer disease is the accumulation of amyloid plaques in the extracellular space in the brain. Amyloid plaques are primarily composed of aggregated amyloid ß peptide (Aß), a proteolytic fragment of the transmembrane amyloid precursor protein (APP). For APP to be proteolytically cleaved into Aß, it must be internalized into the cell and trafficked to endosomes where specific protease complexes can cleave APP. Several recent genome-wide association studies have reported that several single nucleotide polymorphisms (SNPs) in the phosphatidylinositol clathrin assembly lymphoid-myeloid leukemia (PICALM) gene were significantly associated with Alzheimer disease, suggesting a role in APP endocytosis and Aß generation. Here, we show that PICALM co-localizes with APP in intracellular vesicles of N2a-APP cells after endocytosis is initiated. PICALM knockdown resulted in reduced APP internalization and Aß generation. Conversely, PICALM overexpression increased APP internalization and Aß production. In vivo, PICALM was found to be expressed in neurons and co-localized with APP throughout the cortex and hippocampus in APP/PS1 mice. PICALM expression was altered using AAV8 gene transfer of PICALM shRNA or PICALM cDNA into the hippocampus of 6-month-old APP/PS1 mice. PICALM knockdown decreased soluble and insoluble Aß levels and amyloid plaque load in the hippocampus. Conversely, PICALM overexpression increased Aß levels and amyloid plaque load. These data indicate that PICALM, an adaptor protein involved in clathrin-mediated endocytosis, regulates APP internalization and subsequent Aß generation. PICALM contributes to amyloid plaque load in brain likely via its effect on Aß metabolism.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Amiloide/metabolismo , Clatrina/metabolismo , Hipocampo/metabolismo , Proteínas Monoméricas de Montagem de Clatrina/metabolismo , Placa Amiloide/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Amiloide/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Linhagem Celular Tumoral , Clatrina/genética , Técnicas de Silenciamento de Genes , Hipocampo/patologia , Humanos , Camundongos , Proteínas Monoméricas de Montagem de Clatrina/genética , Placa Amiloide/genética , Placa Amiloide/patologia , Transdução Genética
11.
J Neurosci ; 32(13): 4334-40, 2012 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-22457485

RESUMO

Brain region-specific deposition of extracellular amyloid plaques principally composed of aggregated amyloid-ß (Aß) peptide is a pathological signature of Alzheimer's disease (AD). Recent human neuroimaging data suggest that resting-state functional connectivity strength is reduced in patients with AD, cognitively normal elderly harboring elevated amyloid burden, and in advanced aging. Interestingly, there exists a striking spatial correlation between functional connectivity strength in cognitively normal adults and the location of Aß plaque deposition in AD. However, technical limitations have heretofore precluded examination of the relationship between functional connectivity, Aß deposition, and normal aging in mouse models. Using a novel functional connectivity optical intrinsic signal (fcOIS) imaging technique, we demonstrate that Aß deposition is associated with significantly reduced bilateral functional connectivity in multiple brain regions of older APP/PS1 transgenic mice. The amount of Aß deposition in each brain region was associated with the degree of local, age-related bilateral functional connectivity decline. Normal aging was associated with reduced bilateral functional connectivity specifically in retrosplenial cortex. Furthermore, we found that the magnitude of regional bilateral functional correlation in young APP/PS1 mice before Aß plaque formation was proportional to the amount of region-specific plaque deposition seen later in older APP/PS1 mice. Together, these findings suggest that Aß deposition and normal aging are associated with region-specific disruption of functional connectivity and that the magnitude of local bilateral functional connectivity predicts regional vulnerability to subsequent Aß deposition in mouse brain.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/fisiopatologia , Neuroimagem Funcional/estatística & dados numéricos , Vias Neurais/fisiopatologia , Placa Amiloide/metabolismo , Envelhecimento/metabolismo , Envelhecimento/fisiologia , Amiloidose/metabolismo , Amiloidose/fisiopatologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Neuroimagem Funcional/métodos , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Vias Neurais/metabolismo
12.
Science ; 335(6075): 1503-6, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22323736

RESUMO

Alzheimer's disease (AD) is associated with impaired clearance of ß-amyloid (Aß) from the brain, a process normally facilitated by apolipoprotein E (apoE). ApoE expression is transcriptionally induced through the action of the nuclear receptors peroxisome proliferator-activated receptor gamma and liver X receptors in coordination with retinoid X receptors (RXRs). Oral administration of the RXR agonist bexarotene to a mouse model of AD resulted in enhanced clearance of soluble Aß within hours in an apoE-dependent manner. Aß plaque area was reduced more than 50% within just 72 hours. Furthermore, bexarotene stimulated the rapid reversal of cognitive, social, and olfactory deficits and improved neural circuit function. Thus, RXR activation stimulates physiological Aß clearance mechanisms, resulting in the rapid reversal of a broad range of Aß-induced deficits.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Tetra-Hidronaftalenos/farmacologia , Tetra-Hidronaftalenos/uso terapêutico , Amiloidose/tratamento farmacológico , Amiloidose/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Comportamento Animal/efeitos dos fármacos , Bexaroteno , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Líquido Extracelular/efeitos dos fármacos , Líquido Extracelular/metabolismo , Receptores X do Fígado , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Terapia de Alvo Molecular , Odorantes , Condutos Olfatórios/efeitos dos fármacos , Condutos Olfatórios/fisiologia , Receptores Nucleares Órfãos/metabolismo , PPAR gama/metabolismo , Fagocitose , Placa Amiloide/tratamento farmacológico , Receptores X de Retinoides/agonistas , Receptores X de Retinoides/metabolismo
13.
J Alzheimers Dis ; 27(1): 23-38, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21709371

RESUMO

Accumulation of amyloid-ß protein (Aß) in the brain is thought to be a causal event in Alzheimer's disease (AD). Immunotherapy targeting Aß holds great promise for reducing Aß in the brain. Here, we evaluated the efficacy and safety of anti-Aß single-chain antibody (scFv59) delivery via recombinant adeno-associated virus (rAAV) on reducing Aß deposits in an AD mouse model (TgAßPPswe/PS1dE9). First, delivery of scFv59 to the brain was optimized by injecting rAAV serotypes 1, 2, and 5 into the right lateral ventricle. Symmetrical high expression of scFv59 was found throughout the hippocampus and partly in the neocortex in both hemispheres via rAAV1 or rAAV5, while scFv59 expression via rAAV2 was mostly limited to one hemisphere. rAAV1, however, induced apoptosis and microglial activation but rAAV5 did not. Therefore, rAAV5 was selected for therapeutic scFv59 delivery in TgAßPPswe/PS1dE9 mice. rAAV5 was similarly injected into the ventricle of 10-month-old TgAßPPswe/PS1dE9 mice and 5 months later its efficacy and safety were evaluated. Immunoreactive Aß deposits reduced in the hippocampus. Aß42 levels in cerebrospinal fluid (CSF) tended to increase and the Aß40 : 42 ratio decreased in CSF, suggesting that Aß42 was relocated from the parenchyma to CSF. Hemorrhages associated with a focal increase in blood vessel amyloid were found in the brain. While immunotherapy has great potential for clearing cerebral Aß, caution for cerebrovascular effects should be exercised when rAAV-mediated anti-Aß immunotherapy is applied.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/imunologia , Proteínas Amiloidogênicas/metabolismo , Hemorragia Cerebral/induzido quimicamente , Anticorpos de Cadeia Única/efeitos adversos , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Precursor de Proteína beta-Amiloide , Análise de Variância , Animais , Antígenos CD , Apoptose/efeitos dos fármacos , Apoptose/genética , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular Transformada , Dependovirus/fisiologia , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Ensaio de Imunoadsorção Enzimática/métodos , Proteína Glial Fibrilar Ácida/metabolismo , Imunoterapia , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Presenilina-1 , Transdução Genética , Transfecção
14.
Mol Neurodegener ; 4: 51, 2009 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-20017949

RESUMO

BACKGROUND: Recent reports suggest that latrepirdine (Dimebon, dimebolin), a retired Russian antihistamine, improves cognitive function in aged rodents and in patients with mild to moderate Alzheimer's disease (AD). However, the mechanism(s) underlying this benefit remain elusive. AD is characterized by extracellular accumulation of the amyloid-beta (Abeta) peptide in the brain, and Abeta-lowering drugs are currently among the most popular anti-amyloid agents under development for the treatment of AD. In the current study, we assessed the effect of acute dosing of latrepirdine on levels of extracellular Abeta using in vitro and in vivo experimental systems. RESULTS: We evaluated extracellular levels of Abeta in three experimental systems, under basal conditions and after treatment with latrepirdine. Mouse N2a neuroblastoma cells overexpressing Swedish APP were incubated for 6 hr in the presence of either vehicle or vehicle + latrepirdine (500pM-5 muM). Synaptoneurosomes were isolated from TgCRND8 mutant APP-overexpressing transgenic mice and incubated for 0 to 10 min in the absence or presence of latrepirdine (1 muM or 10 muM). Drug-naïve Tg2576 Swedish mutant APP overexpressing transgenic mice received a single intraperitoneal injection of either vehicle or vehicle + latrepirdine (3.5 mg/kg). Picomolar to nanomolar concentrations of acutely administered latrepirdine increased the extracellular concentration of Abeta in the conditioned media from Swedish mutant APP-overexpressing N2a cells by up to 64% (p = 0.01), while a clinically relevant acute dose of latrepirdine administered i.p. led to an increase in the interstitial fluid of freely moving APP transgenic mice by up to 40% (p = 0.01). Reconstitution of membrane protein trafficking and processing is frequently inefficient, and, consistent with this interpretation, latrepirdine treatment of isolated TgCRND8 synaptoneurosomes involved higher concentrations of drug (1-10 muM) and led to more modest increases in extracellular Abeta(x-42 )levels (+10%; p = 0.001); of note, however, was the observation that extracellular Abeta(x-40 )levels did not change. CONCLUSIONS: Here, we report the surprising association of acute latrepirdine dosing with elevated levels of extracellular Abeta as measured in three independent neuron-related or neuron-derived systems, including the hippocampus of freely moving Tg2576 mice. Given the reported association of chronic latrepirdine treatment with improvement in cognitive function, the effects of chronic latrepirdine treatment on extracellular Abeta levels must now be determined.

15.
Neurobiol Dis ; 28(2): 184-96, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17714952

RESUMO

Mice with inactivation of the Tuberous sclerosis complex-1 (Tsc1) gene in glia (Tsc1 GFAP CKO mice) have deficient astrocyte glutamate transporters and develop seizures, suggesting that abnormal glutamate homeostasis contributes to neurological abnormalities in these mice. We examined the hypothesis that Tsc1 GFAP CKO mice have elevated extracellular brain glutamate levels that may cause neuronal death, abnormal glutamatergic synaptic function, and associated impairments in behavioral learning. In vivo microdialysis documented elevated glutamate levels in hippocampi of Tsc1 GFAP CKO mice and several cell death assays demonstrated neuronal death in hippocampus and neocortex. Impairment of long-term potentiation (LTP) with tetanic stimulation was observed in hippocampal slices from Tsc1 GFAP CKO mice and was reversed by low concentrations of NMDA antagonist, indicating that excessive synaptic glutamate directly inhibited LTP. Finally, Tsc1 GFAP CKO mice exhibited deficits in two hippocampal-dependent learning paradigms. These results suggest that abnormal glutamate homeostasis predisposes to excitotoxic cell death, impaired synaptic plasticity and learning deficits in Tsc1 GFAP CKO mice.


Assuntos
Encéfalo/metabolismo , Ácido Glutâmico/metabolismo , Deficiências da Aprendizagem/metabolismo , Plasticidade Neuronal/genética , Transmissão Sináptica/genética , Esclerose Tuberosa/metabolismo , Animais , Astrócitos/metabolismo , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Homeostase/genética , Deficiências da Aprendizagem/genética , Deficiências da Aprendizagem/fisiopatologia , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/genética , Camundongos , Camundongos Knockout , Neocórtex/metabolismo , Neocórtex/fisiopatologia , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Técnicas de Cultura de Órgãos , Esclerose Tuberosa/genética , Esclerose Tuberosa/fisiopatologia , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
16.
Am J Pathol ; 171(1): 241-51, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17591969

RESUMO

Cerebral deposition of the amyloid beta protein (Abeta), an invariant feature of Alzheimer's disease, reflects an imbalance between the rates of Abeta production and clearance. The causes of Abeta elevation in the common late-onset form of Alzheimer's disease (LOAD) are largely unknown. There is evidence that the Abeta-degrading protease neprilysin (NEP) is down-regulated in normal aging and LOAD. We asked whether a decrease in endogenous NEP levels can prolong the half-life of Abeta in vivo and promote development of the classic amyloid neuropathology of Alzheimer's disease. We examined the brains and plasma of young and old mice expressing relatively low levels of human amyloid precursor protein and having one or both NEP genes silenced. NEP loss of function 1) elevated whole-brain and plasma levels of human Abeta(40) and Abeta(42), 2) prolonged the half-life of soluble Abeta in brain interstitial fluid of awake animals, 3) raised the concentration of Abeta dimers, 4) markedly increased hippocampal amyloid plaque burden, and 5) led to the development of amyloid angiopathy. A approximately 50% reduction in NEP levels, similar to that reported in some LOAD brains, was sufficient to increase amyloid neuropathology. These findings demonstrate an important role for proteolysis in determining the levels of Abeta and Abeta-associated neuropathology in vivo and support the hypothesis that primary defects in Abeta clearance can cause or contribute to LOAD pathogenesis.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/irrigação sanguínea , Neprilisina/genética , Placa Amiloide/química , Peptídeos beta-Amiloides/sangue , Animais , Encéfalo/metabolismo , Angiopatia Amiloide Cerebral , Modelos Animais de Doenças , Meia-Vida , Hipocampo/patologia , Humanos , Camundongos , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA