Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 12(8): 769, 2021 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-34349120

RESUMEN

Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder. Increased Aß production plays a fundamental role in the pathogenesis of the disease and BACE1, the protease that triggers the amyloidogenic processing of APP, is a key protein and a pharmacological target in AD. Changes in neuronal activity have been linked to BACE1 expression and Aß generation, but the underlying mechanisms are still unclear. We provide clear evidence for the role of Casein Kinase 2 in the control of activity-driven BACE1 expression in cultured primary neurons, organotypic brain slices, and murine AD models. More specifically, we demonstrate that neuronal activity promotes Casein Kinase 2 dependent phosphorylation of the translation initiation factor eIF4B and this, in turn, controls BACE1 expression and APP processing. Finally, we show that eIF4B expression and phosphorylation are increased in the brain of APPPS1 and APP-KI mice, as well as in AD patients. Overall, we provide a definition of a mechanism linking brain activity with amyloid production and deposition, opening new perspectives from the therapeutic standpoint.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Quinasa de la Caseína II/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Potenciales de Acción , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Quinasa de la Caseína II/antagonistas & inhibidores , Modelos Animales de Enfermedad , Silenciador del Gen , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Presenilina-1/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Regulación hacia Arriba/efectos de los fármacos
2.
Elife ; 102021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33845942

RESUMEN

Previous studies have identified a crucial role of the gut microbiome in modifying Alzheimer's disease (AD) progression. However, the mechanisms of microbiome-brain interaction in AD were so far unknown. Here, we identify microbiota-derived short chain fatty acids (SCFA) as microbial metabolites which promote Aß deposition. Germ-free (GF) AD mice exhibit a substantially reduced Aß plaque load and markedly reduced SCFA plasma concentrations; conversely, SCFA supplementation to GF AD mice increased the Aß plaque load to levels of conventionally colonized (specific pathogen-free [SPF]) animals and SCFA supplementation to SPF mice even further exacerbated plaque load. This was accompanied by the pronounced alterations in microglial transcriptomic profile, including upregulation of ApoE. Despite increased microglial recruitment to Aß plaques upon SCFA supplementation, microglia contained less intracellular Aß. Taken together, our results demonstrate that microbiota-derived SCFA are critical mediators along the gut-brain axis which promote Aß deposition likely via modulation of the microglial phenotype.


Asunto(s)
Ácidos Grasos Volátiles/metabolismo , Microbioma Gastrointestinal , Microglía/metabolismo , Placa Amiloide/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Femenino , Masculino , Ratones , Organismos Libres de Patógenos Específicos
3.
Nat Commun ; 12(1): 1158, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33627648

RESUMEN

Niemann-Pick type C disease is a rare neurodegenerative disorder mainly caused by mutations in NPC1, resulting in abnormal late endosomal/lysosomal lipid storage. Although microgliosis is a prominent pathological feature, direct consequences of NPC1 loss on microglial function remain not fully characterized. We discovered pathological proteomic signatures and phenotypes in NPC1-deficient murine models and demonstrate a cell autonomous function of NPC1 in microglia. Loss of NPC1 triggers enhanced phagocytic uptake and impaired myelin turnover in microglia that precede neuronal death. Npc1-/- microglia feature a striking accumulation of multivesicular bodies and impaired trafficking of lipids to lysosomes while lysosomal degradation function remains preserved. Molecular and functional defects were also detected in blood-derived macrophages of NPC patients that provide a potential tool for monitoring disease. Our study underscores an essential cell autonomous role for NPC1 in immune cells and implies microglial therapeutic potential.


Asunto(s)
Colesterol/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Microglía/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Animales , Western Blotting , Células Cultivadas , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina/metabolismo , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/genética , Fagocitosis/genética , Fagocitosis/fisiología , Proteómica/métodos
4.
EMBO J ; 39(20): e105693, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32954517

RESUMEN

To understand how cells communicate in the nervous system, it is essential to define their secretome, which is challenging for primary cells because of large cell numbers being required. Here, we miniaturized secretome analysis by developing the "high-performance secretome protein enrichment with click sugars" (hiSPECS) method. To demonstrate its broad utility, hiSPECS was used to identify the secretory response of brain slices upon LPS-induced neuroinflammation and to establish the cell type-resolved mouse brain secretome resource using primary astrocytes, microglia, neurons, and oligodendrocytes. This resource allowed mapping the cellular origin of CSF proteins and revealed that an unexpectedly high number of secreted proteins in vitro and in vivo are proteolytically cleaved membrane protein ectodomains. Two examples are neuronally secreted ADAM22 and CD200, which we identified as substrates of the Alzheimer-linked protease BACE1. hiSPECS and the brain secretome resource can be widely exploited to systematically study protein secretion and brain function and to identify cell type-specific biomarkers for CNS diseases.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Oligodendroglía/metabolismo , Proteómica/métodos , Programas Informáticos , Proteínas ADAM/líquido cefalorraquídeo , Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/líquido cefalorraquídeo , Animales , Antígenos CD/líquido cefalorraquídeo , Antígenos CD/metabolismo , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/líquido cefalorraquídeo , Encéfalo/citología , Células Cultivadas , Proteínas del Líquido Cefalorraquídeo , Cromatografía Liquida , Ontología de Genes , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/líquido cefalorraquídeo , Proteínas del Tejido Nervioso/metabolismo , Análisis de Componente Principal , Proteoma/metabolismo , Espectrometría de Masas en Tándem
5.
Elife ; 92020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32510331

RESUMEN

Microglial dysfunction is a key pathological feature of Alzheimer's disease (AD), but little is known about proteome-wide changes in microglia during the course of AD and their functional consequences. Here, we performed an in-depth and time-resolved proteomic characterization of microglia in two mouse models of amyloid ß (Aß) pathology, the overexpression APPPS1 and the knock-in APP-NL-G-F (APP-KI) model. We identified a large panel of Microglial Aß Response Proteins (MARPs) that reflect heterogeneity of microglial alterations during early, middle and advanced stages of Aß deposition and occur earlier in the APPPS1 mice. Strikingly, the kinetic differences in proteomic profiles correlated with the presence of fibrillar Aß, rather than dystrophic neurites, suggesting that fibrillar Aß may trigger the AD-associated microglial phenotype and the observed functional decline. The identified microglial proteomic fingerprints of AD provide a valuable resource for functional studies of novel molecular targets and potential biomarkers for monitoring AD progression or therapeutic efficacy.


Alzheimer's disease is a progressive, irreversible brain disorder. Patients with Alzheimer's have problems with memory and other mental skills, which lead to more severe cognitive decline and, eventually, premature death. This is due to increasing numbers of nerve cells in the brain dying over time. A distinctive feature of Alzheimer's is the abnormally high accumulation of a protein called amyloid-ß, which forms distinctive clumps in the brain termed 'plaques'. The brain has a type of cells called the microglia that identify infections, toxic material and damaged cells, and prevent these from building up by clearing them away. In Alzheimer's disease, however, the microglia do not work properly, which is thought to contribute to the accumulation of amyloid-ß plaques. This means that people with mutations in the genes important for the microglia activity are also at higher risk of developing the disease. Although problems with the microglia play an important role in Alzheimer's, researchers still do not fully understand why microglia stop working in the first place. It is also not known exactly when and how the microglia change as Alzheimer's disease progresses. To unravel this mystery, Sebastian Monasor, Müller et al. carried out a detailed study of the molecular 'fingerprints' of microglia at each key stage of Alzheimer's disease. The experiments used microglia cells from two different strains of genetically altered mice, both of which develop the hallmarks of Alzheimer's disease, including amyloid-ß plaques, at similar rates. Analysis of the proteins in microglia cells from both strains revealed distinctive, large-scale changes corresponding to successive stages of the disease ­ reflecting the gradual accumulation of plaques. Obvious defects in microglia function also appeared soon after plaques started to build up. Microscopy imaging of the brain tissue showed that although amyloid-ß plaques appeared at the same time, they looked different in each mouse strain. In one, plaques were more compact, while in the other, plaques appeared 'fluffier', like cotton wool. In mice with more compacted plaques, microglia recognized the plaques earlier and stopped working sooner, suggesting that plaque structure and microglia defects could be linked. These results shed new light on the role of microglia and their changing protein 'signals' during the different stages of Alzheimer's disease. In the future, this information could help identify people at risk for the disease, so that they can be treated as soon as possible, and to design new therapies to make microglia work again.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Microglía/metabolismo , Proteoma/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Transgénicos , Microglía/patología
6.
EMBO Mol Med ; 11(6)2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31122931

RESUMEN

Microglia adopt numerous fates with homeostatic microglia (HM) and a microglial neurodegenerative phenotype (MGnD) representing two opposite ends. A number of variants in genes selectively expressed in microglia are associated with an increased risk for neurodegenerative diseases such as Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD). Among these genes are progranulin (GRN) and the triggering receptor expressed on myeloid cells 2 (TREM2). Both cause neurodegeneration by mechanisms involving loss of function. We have now isolated microglia from Grn-/- mice and compared their transcriptomes to those of Trem2-/-mice Surprisingly, while loss of Trem2 enhances the expression of genes associated with a homeostatic state, microglia derived from Grn-/- mice showed a reciprocal activation of the MGnD molecular signature and suppression of gene characteristic for HM The opposite mRNA expression profiles are associated with divergent functional phenotypes. Although loss of TREM2 and progranulin resulted in opposite activation states and functional phenotypes of microglia, FDG (fluoro-2-deoxy-d-glucose)-µPET of brain revealed reduced glucose metabolism in both conditions, suggesting that opposite microglial phenotypes result in similar wide spread brain dysfunction.


Asunto(s)
Cerebelo , Glucosa/metabolismo , Glicoproteínas de Membrana/deficiencia , Microglía/metabolismo , Tomografía de Emisión de Positrones , Progranulinas/deficiencia , Receptores Inmunológicos/deficiencia , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Cerebelo/diagnóstico por imagen , Cerebelo/metabolismo , Degeneración Lobar Frontotemporal/diagnóstico por imagen , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/metabolismo , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA