Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 183(6): 1699-1713.e13, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33188775

RESUMEN

To elucidate the role of Tau isoforms and post-translational modification (PTM) stoichiometry in Alzheimer's disease (AD), we generated a high-resolution quantitative proteomics map of 95 PTMs on multiple isoforms of Tau isolated from postmortem human tissue from 49 AD and 42 control subjects. Although Tau PTM maps reveal heterogeneity across subjects, a subset of PTMs display high occupancy and frequency for AD, suggesting importance in disease. Unsupervised analyses indicate that PTMs occur in an ordered manner, leading to Tau aggregation. The processive addition and minimal set of PTMs associated with seeding activity was further defined by analysis of size-fractionated Tau. To summarize, features in the Tau protein critical for disease intervention at different stages of disease are identified, including enrichment of 0N and 4R isoforms, underrepresentation of the C terminus, an increase in negative charge in the proline-rich region (PRR), and a decrease in positive charge in the microtubule binding domain (MBD).


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Procesamiento Proteico-Postraduccional , Proteínas tau/metabolismo , Estudios de Casos y Controles , Estudios de Cohortes , Progresión de la Enfermedad , Humanos , Análisis de Componente Principal , Isoformas de Proteínas/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(12): e2117723119, 2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35290109

RESUMEN

Type 2 diabetes mellitus is known to be a risk factor for Alzheimer's disease (AD), but the underlying mechanisms remain unclear. In AD, the cerebral accumulation of amyloid ß (Aß) triggers a pathological cascade leading to neurodegeneration. Plasma Aß levels are thought to reflect the brain amyloid pathology and currently used as a diagnostic biomarker of AD. However, amyloid precursor protein and Aß-generating enzymes, ß- and γ-secretases, are widely expressed in various peripheral tissues. Previous reports have shown that glucose and insulin loading cause a transient increase of plasma Aß in mice and humans. These findings led us to speculate that plasma Aß is produced from glucose- and insulin-susceptible peripheral tissues to play a role in glucose and insulin metabolism. To test this hypothesis, we investigated the effects of glucose and insulin on Aß secretion and the effect of Aß on insulin secretion in vivo, ex vivo, and in vitro. Aß was found to be secreted from ß-cells of the pancreas along with insulin upon glucose stimulation. Upon insulin stimulation, Aß was secreted from cells of insulin-targeted organs, such as adipose tissues, skeletal muscles, and the liver, along with their organokines. Furthermore, Aß inhibited the glucose-triggered insulin secretion from ß-cells, slowing down glucose clearance from the blood. These results suggest that peripheral Aß acts as a negative modulator of insulin secretion. Our findings provide a possible mechanism linking diabetes to AD and call attention to how plasma Aß levels are used in AD diagnosis.


Asunto(s)
Enfermedad de Alzheimer , Diabetes Mellitus Tipo 2 , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina
3.
Biochem Biophys Res Commun ; 721: 150025, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38768546

RESUMEN

The causes of Alzheimer's disease (AD) are poorly understood, although many genes are known to be involved in this pathology. To gain insights into the underlying molecular mechanisms, it is essential to identify the relationships between individual AD genes. Previous work has shown that the splice variant E of KLC1 (KLC1_vE) promotes AD, and that the CELF1 gene, which encodes an RNA-binding protein involved in splicing regulation, is at a risk locus for AD. Here, we identified a functional link between CELF1 and KLC1 in AD pathogenesis. Transcriptomic data from human samples from different ethnic groups revealed that CELF1 mRNA levels are low in AD brains, and the splicing pattern of KLC1 is strongly correlated with CELF1 expression levels. Specifically, KLC1_vE is negatively correlated with CELF1. Depletion and overexpression experiments in cultured cells demonstrated that the CELF1 protein down-regulates KLC1_vE. In a cross-linking and immunoprecipitation sequencing (CLIP-seq) database, CELF1 directly binds to KLC1 RNA, following which it likely modulates terminal exon usage, hence KLC1_vE formation. These findings reveal a new pathogenic pathway where a risk allele of CELF1 is associated with reduced CELF1 expression, which up-regulates KLC1_vE to promote AD.


Asunto(s)
Empalme Alternativo , Enfermedad de Alzheimer , Proteínas CELF1 , Humanos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Proteínas CELF1/metabolismo , Proteínas CELF1/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética
4.
FASEB J ; 36(10): e22555, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36125010

RESUMEN

Although α-synuclein (SNCA) is a well-known pathological molecule involved in synucleinopathy in neurons, its physiological roles remain largely unknown. We reported that serum SNCA levels have a close inverse correlation with blood pressure and age, which indicates the involvement of SNCA in age-related endothelial dysfunction. Therefore, this study aimed to elucidate the molecular functions of SNCA in the endothelium. We confirmed that SNCA was expressed in and secreted from endothelial cells (ECs). Exogenous treatment with recombinant SNCA (rSNCA) activated the Akt-eNOS axis and increased nitric oxide production in ECs. Treatment with rSNCA also suppressed TNF-α- and palmitic acid-induced NF-κB activation, leading to the suppression of VCAM-1 upregulation and restoration of eNOS downregulation in ECs. As for endogenous SNCA expression, replicative senescence resulted in the attenuation of SNCA expression in cultured ECs, similar to the effects of physiological aging on mice aortas. The siRNA-mediated silencing of SNCA consistently resulted in senescent phenotypes, such as eNOS downregulation, increased ß-gal activity, decreased Sirt1 expression, and increased p53 expression, in ECs. Ex vivo assessment of endothelial functions using aortic rings revealed impaired endothelium-dependent acetylcholine-induced relaxation in SNCA knockout (KO) mice. Furthermore, SNCA KO mice, especially those on a high-fat diet, displayed elevated blood pressure compared with wild-type mice; this could be eNOS dysfunction-dependent because of the lower difference caused by L-NAME administration. These results indicate that exogenous and endogenous SNCA in ECs might physiologically maintain vascular integrity, and age-related endothelial dysfunction might be partially ascribed to loss-of-function of SNCA in ECs.


Asunto(s)
Enfermedades Vasculares , alfa-Sinucleína/metabolismo , Acetilcolina/metabolismo , Animales , Células Endoteliales/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Ácido Palmítico/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Sirtuina 1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Enfermedades Vasculares/metabolismo
5.
Dement Geriatr Cogn Disord ; 52(2): 108-116, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36878194

RESUMEN

INTRODUCTION: A rapidly increasing number of patients with dementia present a serious social problem. Recently, the incidence of epilepsy in patients with Alzheimer's disease (AD) is increasing, drawing attention to the pathological relationship between the two conditions. Clinical studies have suggested the protective action of antiepileptic agents on dementia; however, the underlying mechanism remains unknown. We evaluated the effects of multiple antiepileptic drugs using tau aggregation assay systems to determine the effects of antiepileptic agents on tau aggregation, a major neuropathological finding associated with AD. METHODS: We evaluated the effects of seven antiepileptic agents on intracellular tau aggregation using a tau-biosensor cell-based high-throughput assay. Next, we tested these agents in a cell-free tau aggregation assay using thioflavin T (ThT). RESULTS: The assay results revealed that phenobarbital inhibited tau aggregation, whereas sodium valproate, gabapentin, and piracetam promoted tau aggregation. In the cell-free tau aggregation assay using ThT, we confirmed that phenobarbital significantly inhibited tau aggregation. CONCLUSION: Antiepileptic drugs may modify the tau pathology in AD in a neural activity-independent manner. Our finding may provide an important insight into the optimization of antiepileptic drug therapy in older adults with dementia.


Asunto(s)
Enfermedad de Alzheimer , Anticonvulsivantes , Humanos , Anciano , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Proteínas tau , Ácido Valproico/farmacología , Ácido Valproico/uso terapéutico , Fenobarbital/uso terapéutico
6.
FASEB J ; 34(2): 2425-2435, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31907998

RESUMEN

Clinical studies have indicated that obesity and diabetes are associated with Alzheimer's disease (AD) and neurodegeneration. Although the mechanisms underlying these associations remain elusive, the bidirectional interactions between obesity/diabetes and Alzheimer's disease (AD) may be involved in them. Both obesity/diabetes and AD significantly reduce life expectancy. We generated AppNL-F/wt knock-in; ob/ob mice by crossing AppNL-F/wt knock-in mice and ob/ob mice to investigate whether amyloid-ß (Aß) affects the lifespan of ob/ob mice. AppNL-F/wt knock-in; ob/ob mice displayed the shortest lifespan compared to wild-type mice, AppNL-F/wt knock-in mice, and ob/ob mice. Notably, the Aß42 levels were increased at minimum levels before deposition in AppNL-F/wt knock-in mice and AppNL-F/wt knock-in; ob/ob mice at 18 months of age. No differences in the levels of several neuronal markers were observed between mice at this age. However, we observed increased levels of glial fibrillary acidic protein (GFAP), an astrocyte marker, in AppNL-F/wt knock-in; ob/ob mice, while the levels of several microglial markers, including CD11b, TREM2, and DAP12, were decreased in both ob/ob mice and AppNL-F/wt knock-in; ob/ob mice. The increase in GFAP levels was not observed in young AppNL-F/wt knock-in; ob/ob mice. Thus, the increased Aß42 levels may decrease the lifespan of ob/ob mice, which is associated with the dysregulation of microglia and astrocytes in an age-dependent manner. Based on these findings, the imbalance in these neuroinflammatory cells may provide a clue to the mechanisms by which the interaction between obesity/diabetes and early AD reduces life expectancy.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Astrocitos/metabolismo , Longevidad , Microglía/metabolismo , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Astrocitos/patología , Técnicas de Sustitución del Gen , Ratones , Ratones Noqueados , Ratones Obesos , Microglía/patología , Fragmentos de Péptidos/genética
7.
FASEB J ; 34(2): 2792-2811, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31912559

RESUMEN

While adipose tissue is required to maintain glucose metabolism, excessive calorie intake induces obesity via mechanisms including accelerated proliferation and differentiation of preadipocytes, leading to insulin resistance. Here, we investigated the role of myoferlin (MYOF), a ferlin family protein, in regulating glucose metabolism by mainly focusing on its unknown role in adipose tissue. Whereas young MYOF knockout (KO) mice on a normal diet showed aggravated glucose tolerance and insulin sensitivity, those on a high-fat diet (HFD) showed preserved glucose tolerance with an attenuated gain of body weight, reduced visceral fat deposits, and less severe fatty liver. The Adipose MYOF expression was reduced by aging but was restored by an HFD along with the retained expression of NFAT transcription factors. Loss-of-function of MYOF in preadipocytes suppressed proliferation and differentiation into mature adipocytes along with the decreased expression of genes involved in adipogenesis. The MYOF expression in preadipocytes was reduced with differentiation. Attenuated obesity in MYOF KO mice on an HFD was also accompanied with increased oxygen consumption by an unidentified mechanism and with reduced adipose inflammation due to less inflammatory macrophages. These insights suggest that the multifunctional roles of MYOF involve the regulation of preadipocyte function and affect glucose metabolism bidirectionally depending on consumed calories.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis/fisiología , Adiposidad/fisiología , Glucosa/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Musculares/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Diferenciación Celular , Inflamación/metabolismo , Resistencia a la Insulina/fisiología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL
8.
EMBO J ; 34(24): 3028-41, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26538322

RESUMEN

In Alzheimer's disease and tauopathies, tau protein aggregates into neurofibrillary tangles that progressively spread to synaptically connected brain regions. A prion-like mechanism has been suggested: misfolded tau propagating through the brain seeds neurotoxic aggregation of soluble tau in recipient neurons. We use transgenic mice and viral tau expression to test the hypotheses that trans-synaptic tau propagation, aggregation, and toxicity rely on the presence of endogenous soluble tau. Surprisingly, mice expressing human P301Ltau in the entorhinal cortex showed equivalent tau propagation and accumulation in recipient neurons even in the absence of endogenous tau. We then tested whether the lack of endogenous tau protects against misfolded tau aggregation and toxicity, a second prion model paradigm for tau, using P301Ltau-overexpressing mice with severe tangle pathology and neurodegeneration. Crossed onto tau-null background, these mice had similar tangle numbers but were protected against neurotoxicity. Therefore, misfolded tau can propagate across neural systems without requisite templated misfolding, but the absence of endogenous tau markedly blunts toxicity. These results show that tau does not strictly classify as a prion protein.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas tau/genética , Animales , Células Cultivadas , Corteza Entorrinal/citología , Corteza Entorrinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación Missense , Neuronas/metabolismo , Proteínas tau/deficiencia , Proteínas tau/metabolismo
9.
Am J Pathol ; 187(6): 1399-1412, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28408124

RESUMEN

The clinical progression of Alzheimer disease (AD) is associated with the accumulation of tau neurofibrillary tangles, which may spread throughout the cortex by interneuronal tau transfer. If so, targeting extracellular tau species may slow the spreading of tau pathology and possibly cognitive decline. To identify suitable target epitopes, we tested the effects of a panel of tau antibodies on neuronal uptake and aggregation in vitro. Immunodepletion was performed on brain extract from tau-transgenic mice and postmortem AD brain and added to a sensitive fluorescence resonance energy transfer-based tau uptake assay to assess blocking efficacy. The antibodies reduced tau uptake in an epitope-dependent manner: N-terminal (Tau13) and middomain (6C5 and HT7) antibodies successfully prevented uptake of tau species, whereas the distal C-terminal-specific antibody (Tau46) had little effect. Phosphorylation-dependent (40E8 and p396) and C-terminal half (4E4) tau antibodies also reduced tau uptake despite removing less total tau by immunodepletion, suggesting specific interactions with species involved in uptake. Among the seven antibodies evaluated, 6C5 most efficiently blocked uptake and subsequent aggregation. More important, 6C5 also blocked neuron-to-neuron spreading of tau in a unique three-chamber microfluidic device. Furthermore, 6C5 slowed down the progression of tau aggregation even after uptake had begun. Our results imply that not all antibodies/epitopes are equally robust in terms of blocking tau uptake of human AD-derived tau species.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Neuronas/metabolismo , Proteínas tau/metabolismo , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Epítopos/inmunología , Femenino , Humanos , Interneuronas/metabolismo , Masculino , Ratones Transgénicos , Técnicas Analíticas Microfluídicas , Terapia Molecular Dirigida/métodos , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Fosforilación , Proteínas tau/antagonistas & inhibidores , Proteínas tau/inmunología
10.
Cell Mol Neurobiol ; 38(8): 1539-1550, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30288631

RESUMEN

In Parkinson's disease and other Lewy body disorders, the propagation of pathology has been accredited to the spreading of extracellular α-synuclein (α-syn). Although the pathogenic mechanisms are not fully understood, cell-to-cell transfer of α-syn via exosomes and other extracellular vesicles (EVs) has been reported. Here, we investigated whether altered molecular properties of α-syn can influence the distribution and secretion of α-syn in human neuroblastoma cells. Different α-syn variants, including α-syn:hemi-Venus and disease-causing mutants, were overexpressed and EVs were isolated from the conditioned medium. Of the secreted α-syn, 0.1-2% was associated with vesicles. The major part of EV α-syn was attached to the outer membrane of vesicles, whereas a smaller fraction was found in their lumen. For α-syn expressed with N-terminal hemi-Venus, the relative levels associated with EVs were higher than for WT α-syn. Moreover, such EV-associated α-syn:hemi-Venus species were internalized in recipient cells to a higher degree than the corresponding free-floating forms. Among the disease-causing mutants, A53T α-syn displayed an increased association with EVs. Taken together, our data suggest that α-syn species with presumably lost physiological functions or altered aggregation properties may shift the cellular processing towards vesicular secretion. Our findings thus lend further support to the tenet that EVs can mediate spreading of harmful α-syn species and thereby contribute to the pathology in α-synucleinopathies.


Asunto(s)
Vesículas Extracelulares/metabolismo , alfa-Sinucleína/metabolismo , Biomarcadores/metabolismo , Células Cultivadas , Exosomas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas Mutantes/metabolismo , Proteínas tau/metabolismo
11.
Ann Neurol ; 80(3): 355-67, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27351289

RESUMEN

OBJECTIVE: Cerebrospinal fluid (CSF) tau is an excellent surrogate marker for assessing neuropathological changes that occur in Alzheimer's disease (AD) patients. However, whether the elevated tau in AD CSF is just a marker of neurodegeneration or, in fact, a part of the disease process is uncertain. Moreover, it is unknown how CSF tau relates to the recently described soluble high-molecular-weight (HMW) species that is found in the postmortem AD brain and can be taken up by neurons and seed aggregates. METHODS: We have examined seeding and uptake properties of brain extracellular tau from various sources, including interstitial fluid (ISF) and CSF from an AD transgenic mouse model and postmortem ventricular and antemortem lumbar CSF from AD patients. RESULTS: We found that brain ISF and CSF tau from the AD mouse model can be taken up by cells and induce intracellular aggregates. Ventricular CSF from AD patients contained a rare HMW tau species that exerted a higher seeding activity. Notably, the HMW tau species was also detected in lumbar CSF from AD patients, and its levels were significantly elevated compared to control subjects. HMW tau derived from CSF of AD patients was seed competent in vitro. INTERPRETATION: These findings suggest that CSF from an AD brain contains potentially bioactive HMW tau species, giving new insights into the role of CSF tau and biomarker development for AD. Ann Neurol 2016;80:355-367.


Asunto(s)
Enfermedad de Alzheimer/líquido cefalorraquídeo , Encéfalo/metabolismo , Proteínas tau/líquido cefalorraquídeo , Anciano , Animales , Biomarcadores/líquido cefalorraquídeo , Líquido Extracelular/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad
12.
J Biol Chem ; 290(4): 1966-78, 2015 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-25468905

RESUMEN

Interfering with the assembly of Amyloid ß (Aß) peptides from monomer to oligomeric species and fibrils or promoting their clearance from the brain are targets of anti-Aß-directed therapies in Alzheimer disease. Here we demonstrate that cromolyn sodium (disodium cromoglycate), a Food and Drug Administration-approved drug already in use for the treatment of asthma, efficiently inhibits the aggregation of Aß monomers into higher-order oligomers and fibrils in vitro without affecting Aß production. In vivo, the levels of soluble Aß are decreased by over 50% after only 1 week of daily intraperitoneally administered cromolyn sodium. Additional in vivo microdialysis studies also show that this compound decreases the half-life of soluble Aß in the brain. These data suggest a clear effect of a peripherally administered, Food and Drug Administration-approved medication on Aß economy, supporting further investigation of the potential long-term efficacy of cromolyn sodium in Alzheimer disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Cromolin Sódico/farmacología , Aprobación de Drogas , Fragmentos de Péptidos/metabolismo , Animales , Células Cultivadas , Cromolin Sódico/química , Modelos Animales de Enfermedad , Flavonoides/química , Flavonoles , Humanos , Ratones , Ratones Transgénicos , Microglía/metabolismo , Microscopía Electrónica de Transmisión , Estados Unidos , United States Food and Drug Administration
13.
J Neurochem ; 139(6): 1163-1174, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27731899

RESUMEN

Tau is a neuronal microtubule-binding protein that, in Alzheimer's disease and other neurodegenerative diseases, can form oligomeric and large fibrillar aggregates, which deposit in neurofibrillary tangles. Tau's physiological state of multimerization appears to vary across conditions, and a stable dimeric form of soluble tau has been suggested from experiments using recombinant tau in vitro. We tested if tau dimerization or oligomerization, also occurs in cells, and if soluble tau oligomers are relevant for the release and internalization of tau. We developed a sensitive tau split-luciferase assay to show the rapid intracellular formation of stable tau dimers that are released and taken up by cells. Our data further suggest that tau dimerization can be accelerated slightly by aggregation catalysts. We conclude that tau oligomers are a stable physiological form of tau, and that tau oligomerization does not necessarily lead to tau aggregation.


Asunto(s)
Multimerización de Proteína/fisiología , Proteínas tau/metabolismo , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Ovillos Neurofibrilares/metabolismo , Unión Proteica/fisiología , Proteínas tau/toxicidad
14.
Neurobiol Dis ; 84: 109-19, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26102023

RESUMEN

Oxidative stress is a common feature of the aging process and of many neurodegenerative disorders, including Alzheimer's disease. Understanding the direct causative relationship between oxidative stress and amyloid pathology, and determining the underlying molecular mechanisms is crucial for the development of more effective therapeutics for the disease. By employing microdialysis technique, we report local increase in the amyloid-ß42 levels and elevated amyloid-ß42/40 ratio in the interstitial fluid within 6h of direct infusion of oxidizing agents into the hippocampus of living and awake wild type mice. The increase in the amyloid-ß42/40 ratio correlated with the pathogenic conformational change of the amyloid precursor protein-cleaving enzyme, presenilin1/γ-secretase. Furthermore, we found that the product of lipid peroxidation 4-hydroxynonenal, binds to both nicastrin and BACE, differentially affecting γ- and ß-secretase activity, respectively. The present study demonstrates a direct cause-and-effect correlation between oxidative stress and altered amyloid-ß production, and provides a molecular mechanism by which naturally occurring product of lipid peroxidation may trigger generation of toxic amyloid-ß42 species.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Peroxidación de Lípido/fisiología , Estrés Oxidativo/fisiología , Fragmentos de Péptidos/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Aldehídos/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Antioxidantes/farmacología , Encéfalo/efectos de los fármacos , Disulfuros/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Ratones , Estrés Oxidativo/efectos de los fármacos , Presenilina-1/metabolismo , Piridinas/metabolismo
15.
FASEB J ; 27(8): 3239-48, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23640054

RESUMEN

There is a growing body of evidence that soluble oligomeric forms of amyloid ß (Aß) play a critical role in Alzheimer's disease (AD). Despite the importance of soluble Aß oligomers as a therapeutic target for AD, the dynamic metabolism of these Aß species in vivo has not been elucidated because of the difficulty in monitoring brain Aß oligomers in living animals. Here, using a unique large pore-sized membrane microdialysis, we characterized soluble Aß oligomers in brain interstitial fluid (ISF) of awake, freely moving APP/PS1 transgenic and control WT mice. We could detect high-molecular-weight (HMW) and low-molecular-weight (LMW) Aß oligomers in the brain ISF of living animals, which increased dramatically in an age-dependent manner (5- to 8-fold increase, 4 vs. 17-18 mo). Notably, HMW Aß decreased more slowly than other forms of Aß after acute γ-secretase inhibition [% decrease from the baseline (HMW vs. LMW) was 36.9 vs. 74.1% (Aß40, P<0.05) and 25.4 vs. 88.0% (Aß42, P<0.01)], suggesting that HMW Aß oligomers clear more slowly than other forms from the brain. These data reveal the dynamic metabolism of neurotoxic Aß oligomers in AD brain and could provide new insights into Aß-targeted therapies for AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Factores de Edad , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/química , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Benzodiazepinonas/farmacología , Encéfalo/efectos de los fármacos , Líquido Extracelular/efectos de los fármacos , Líquido Extracelular/metabolismo , Humanos , Immunoblotting , Ratones , Ratones Transgénicos , Microdiálisis/métodos , Peso Molecular , Presenilina-1/genética , Presenilina-1/metabolismo , Multimerización de Proteína
16.
AJNR Am J Neuroradiol ; 45(3): 320-327, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38331963

RESUMEN

BACKGROUND AND PURPOSE: Biomarkers have been required for diagnosing early Alzheimer disease. We assessed the utility of hippocampal diffusion parameters for diagnosing Alzheimer disease pathology in mild cognitive impairment. MATERIALS AND METHODS: Sixty-nine patients with mild cognitive impairment underwent both CSF measurement and multi-shell diffusion imaging at 3T. Based on the CSF biomarker level, patients were classified according to the presence (Alzheimer disease group, n = 35) or absence (non-Alzheimer disease group, n = 34) of Alzheimer disease pathology. Neurite orientation dispersion and density imaging and diffusion tensor imaging parametric maps were generated. Two observers independently created the hippocampal region of interest for calculating histogram features. Interobserver correlations were calculated. The statistical significance of intergroup differences was tested by using the Mann-Whitney U test. Logistic regression analyses, using both the clinical scale and the image data, were used to predict intergroup differences, after which group discriminations were performed. RESULTS: Most intraclass correlation coefficient values were between 0.59 and 0.91. In the regions of interest of both observers, there were statistically significant intergroup differences for the left-side neurite orientation dispersion and density imaging-derived intracellular volume fraction, right-side diffusion tensor imaging-derived mean diffusivity, left-side diffusion tensor imaging-derived mean diffusivity, axial diffusivity, and radial diffusivity (P < .05). Logistic regression models revealed that diffusion parameters contributed the most to discriminating between the groups. The areas under the receiver operating characteristic curve for the regions of interest of observers A/B were 0.69/0.68, 0.69/0.68, 0.73/0.68, 0.71/0.68, and 0.68/0.68 for the left-side intracellular volume fraction (mean), right-side mean diffusivity (mean), left-side mean diffusivity (10th percentile), axial diffusivity (10th percentile), and radial diffusivity (mean). CONCLUSIONS: Hippocampal diffusion parameters might be useful for the early diagnosis of Alzheimer disease.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/patología , Imagen de Difusión Tensora/métodos , Imagen de Difusión por Resonancia Magnética/métodos , Hipocampo/diagnóstico por imagen , Hipocampo/patología , Biomarcadores
17.
J Neurosci ; 32(43): 15181-92, 2012 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-23100439

RESUMEN

Alzheimer's disease (AD) is the most common progressive neurodegenerative disorder causing dementia. Massive deposition of amyloid ß peptide (Aß) as senile plaques in the brain is the pathological hallmark of AD, but oligomeric, soluble forms of Aß have been implicated as the synaptotoxic component. The apolipoprotein E ε 4 (apoE ε4) allele is known to be a genetic risk factor for developing AD. However, it is still unknown how apoE impacts the process of Aß oligomerization. Here, we found that the level of Aß oligomers in APOE ε4/ε4 AD patient brains is 2.7 times higher than those in APOE ε3/ε3 AD patient brains, matched for total plaque burden, suggesting that apoE4 impacts the metabolism of Aß oligomers. To test this hypothesis, we examined the effect of apoE on Aß oligomer formation. Using both synthetic Aß and a split-luciferase method for monitoring Aß oligomers, we observed that apoE increased the level of Aß oligomers in an isoform-dependent manner (E2 < E3 < E4). This effect appears to be dependent on the ApoE C-terminal domain. Moreover, these results were confirmed using endogenous apoE isolated from the TBS-soluble fraction of human brain, which increased the formation of Aß oligomers. Together, these data show that lipidated apoE, especially apoE4, increases Aß oligomers in the brain. Higher levels of Aß oligomers in the brains of APOE ε4/ε4 carriers compared with APOE ε3/ε3 carriers may increase the loss of dendritic spines and accelerate memory impairments, leading to earlier cognitive decline in AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Encéfalo/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/farmacología , Análisis de Varianza , Apolipoproteína E2/genética , Apolipoproteína E2/metabolismo , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Astrocitos/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Proteínas Fluorescentes Verdes/genética , Células HEK293/efectos de los fármacos , Células HEK293/metabolismo , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Masculino , Morfolinos/farmacología , Fragmentos de Péptidos/farmacología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transfección
18.
Proc Natl Acad Sci U S A ; 107(15): 7036-41, 2010 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-20231468

RESUMEN

Recent epidemiological studies suggest that diabetes mellitus is a strong risk factor for Alzheimer disease. However, the underlying mechanisms remain largely unknown. In this study, to investigate the pathophysiological interaction between these diseases, we generated animal models that reflect the pathologic conditions of both diseases. We crossed Alzheimer transgenic mice (APP23) with two types of diabetic mice (ob/ob and NSY mice), and analyzed their metabolic and brain pathology. The onset of diabetes exacerbated Alzheimer-like cognitive dysfunction without an increase in brain amyloid-beta burden in double-mutant (APP(+)-ob/ob) mice. Notably, APP(+)-ob/ob mice showed cerebrovascular inflammation and severe amyloid angiopathy. Conversely, the cross-bred mice showed an accelerated diabetic phenotype compared with ob/ob mice, suggesting that Alzheimer amyloid pathology could aggravate diabetes. Similarly, APP(+)-NSY fusion mice showed more severe glucose intolerance compared with diabetic NSY mice. Furthermore, high-fat diet feeding induced severe memory deficits in APP(+)-NSY mice without an increase in brain amyloid-beta load. Here, we created Alzheimer mouse models with early onset of cognitive dysfunction. Cerebrovascular changes and alteration in brain insulin signaling might play a pivotal role in this relationship. These findings could provide insights into this intensely debated association.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/biosíntesis , Diabetes Mellitus Experimental/fisiopatología , Trastornos de la Memoria/fisiopatología , Enfermedad de Alzheimer/complicaciones , Alimentación Animal , Animales , Circulación Cerebrovascular , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Femenino , Inflamación , Insulina/metabolismo , Masculino , Trastornos de la Memoria/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
19.
Sci Adv ; 9(47): eadg3193, 2023 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-37992159

RESUMEN

Repulsive guidance molecule A (RGMa) was originally identified as a neuronal growth cone-collapsing factor. Previous reports have demonstrated the multifunctional roles of RGMa mediated by neogenin1. However, the pathogenic involvement of RGMa in amyotrophic lateral sclerosis (ALS) remains unclear. Here, we demonstrated that RGMa concentration was elevated in the cerebrospinal fluid of both patients with ALS and transgenic mice overexpressing the mutant human superoxide dismutase1 (mSOD1 mice). Treatment with humanized anti-RGMa monoclonal antibody ameliorated the clinical symptoms in mSOD1 mice. Histochemical analysis revealed that the anti-RGMa antibody significantly decreased mutant SOD1 protein accumulation in the motor neurons of mSOD1 mice via inhibition of actin depolymerization. In vitro analysis revealed that the anti-RGMa antibody inhibited the cellular uptake of the mutant SOD1 protein, presumably by reinforcing the neuronal actin barrier. Collectively, these data suggest that RGMa leads to the collapse of the neuronal actin barrier and promotes aberrant protein deposition, resulting in exacerbation of the ALS pathology.


Asunto(s)
Esclerosis Amiotrófica Lateral , Animales , Humanos , Ratones , Actinas , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Anticuerpos , Ratones Transgénicos , Neuronas Motoras/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/genética
20.
Dement Geriatr Cogn Disord ; 34(1): 25-30, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22889768

RESUMEN

BACKGROUND: Although plasma ß-amyloid (Aß) has been suggested to be a noninvasive diagnostic biomarker for Alzheimer's disease (AD), its significance and validity have been inconclusive. Thus, it is quite important to establish a novel diagnostic method related to plasma Aß. METHODS: As our previous animal studies demonstrated a relation of glucose with plasma Aß, we examined the effect of glucose loading on plasma Aß levels in AD patients. After fasting, an oral glucose load was administered to AD patients and non-AD dementia patients, and subsequently, blood glucose, plasma insulin, and plasma Aß levels were measured. RESULTS: The plasma levels of baseline blood glucose, plasma insulin, and plasma Aß were not different between the two groups. However, immediately after glucose loading, a significant increase in plasma Aß40 and Aß42 levels was observed in AD patients, whereas a mild decrease in plasma Aß40 and Aß42 levels was detected in non-AD dementia patients. CONCLUSION: The present study clearly demonstrated a different response in plasma Aß40 and Aß42 levels after glucose loading between AD and non-AD dementia patients, which is consistent with our previous animal studies. These findings suggest a novel diagnostic tool for AD using the elevation of plasma Aß level after glucose loading, although further studies are necessary.


Asunto(s)
Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/diagnóstico , Péptidos beta-Amiloides/sangre , Glucosa , Anciano , Anciano de 80 o más Años , Área Bajo la Curva , Biomarcadores/sangre , Glucemia/análisis , Demencia Vascular/sangre , Demencia Vascular/diagnóstico , Diagnóstico Diferencial , Femenino , Demencia Frontotemporal/sangre , Demencia Frontotemporal/diagnóstico , Humanos , Hidrocéfalo Normotenso/sangre , Hidrocéfalo Normotenso/inducido químicamente , Insulina/sangre , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA