Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Immunity ; 47(3): 398-400, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28930654

RESUMEN

Microglial cell function is implicated in the etiology of Alzheimer's disease by human genetics. In this issue of Immunity, Krasemann et al. (2017) describe a gene expression signature associated with an APOE- and TREM2-dependent response of microglia to brain tissue damage that accumulates in aging and disease, defining an axis that might be amenable to therapeutic targeting.


Asunto(s)
Apolipoproteínas E , Microglía/inmunología , Envejecimiento , Enfermedad de Alzheimer , Encéfalo , Humanos
2.
Neurobiol Dis ; 148: 105217, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33301878

RESUMEN

More than forty loci contribute to genetic risk for Alzheimer's disease (AD). These risk alleles are enriched in myeloid cell enhancers suggesting that microglia, the brain-resident macrophages, contribute to AD risk. We have previously identified SPI1/PU.1, a master regulator of myeloid cell development in the brain and periphery, as a genetic risk factor for AD. Higher expression of SPI1 is associated with increased risk for AD, while lower expression is protective. To investigate the molecular and cellular phenotypes associated with higher and lower expression of PU.1 in microglia, we used stable overexpression and knock-down of PU.1 in BV2, an immortalized mouse microglial cell line. Transcriptome analysis suggests that reduced PU.1 expression suppresses expression of homeostatic genes similar to the disease-associated microglia response to amyloid plaques in mouse models of AD. Moreover, PU.1 knock-down resulted in activation of protein translation, antioxidant action and cholesterol/lipid metabolism pathways with a concomitant decrease of pro-inflammatory gene expression. PU.1 overexpression upregulated and knock-down downregulated phagocytic uptake in BV2 cells independent of the nature of the engulfed material. However, cells with reduced PU.1 expression retained their ability to internalize myelin similar to control albeit with a delay, which aligns with their anti-inflammatory profile. Here we identified several microglial responses that are modulated by PU.1 expression levels and propose that risk association of PU.1 to AD is driven by increased pro-inflammatory response due to increased viability of cells under cytotoxic conditions. In contrast, low expression of PU.1 leads to increased cell death under cytotoxic conditions accompanied by reduced pro-inflammatory signaling that decreased A1 reactive astrocytes signature supporting the protective effect of SPI1 genotype in AD. These findings inform future in vivo validation studies and design of small molecule screens for therapeutic discovery in AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Apoptosis/genética , Inflamación/genética , Microglía/metabolismo , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Ratones , Microglía/efectos de los fármacos , Óxido Nítrico/metabolismo , Fragmentos de Péptidos/farmacología , Rotenona/farmacología , Estaurosporina , Desacopladores/farmacología
3.
Neurobiol Dis ; 138: 104785, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32032730

RESUMEN

Mutations in APP (amyloid precursor protein), PSEN1 (presenilin 1) or PSEN2 (presenilin 2) are the main cause of early-onset familial forms of Alzheimer's disease (autosomal dominant AD or ADAD). These genes affect γ-secretase-dependent generation of Amyloid ß (Aß) peptides, the main constituent of amyloid plaques and one of the pathological hallmarks of AD. Evaluation of patients with ADAD includes assessment of family history, clinical presentation, biomarkers, neuropathology when available and DNA sequencing data. These analyses frequently uncover novel variants of unknown significance in ADAD genes. This presents a barrier to recruitment of such individuals into clinical trials, unless a biochemical test can demonstrate that a novel mutation results in altered APP processing in a manner consistent with pathogenicity. Here we describe generation and characterization of a novel presenilin 1 and 2 double knock-out in N2A mouse neuroblastoma cells using CRISPR/Cas9, which results in complete ablation of Aß production, decreased Pen-2 expression and Nicastrin glycosylation. Because of the absence of background Aß secretion from endogenous γ-secretases, these cells can be used for validation of PSEN1 and PSEN2 variant effects on production of Aß or other γ-secretase substrates and for biochemical studies of γ-secretase function using novel variants. We examined several PSEN1 and PSEN2 mutations of known and unknown pathogenicity. Known mutants increased Aß42/Aß40 ratio with varying effect on Aß40, Aß42, total Aß levels and Pen-2 expression, which aligns with previous work on these mutants. Our data on novel PSEN1 V142F, G206V and G206D mutations suggest that these mutations underlie the reported clinical observations in ADAD patients. We believe our novel cell line will be valuable for the scientific community for reliable validation of presenilin mutations and helpful in defining their pathogenicity to improve and facilitate evaluation of ADAD patients, particularly in the context of enrollment in clinical trials.


Asunto(s)
Enfermedad de Alzheimer/genética , Presenilina-1/genética , Presenilina-2/genética , Enfermedad de Alzheimer/diagnóstico , Secretasas de la Proteína Precursora del Amiloide/genética , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Línea Celular , Ratones , Mutación , Selección de Paciente , Fragmentos de Péptidos/metabolismo , Placa Amiloide/genética
4.
Neurobiol Dis ; 139: 104817, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32087291

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disease that is clinically characterized by progressive cognitive decline. More than 200 pathogenic mutations have been identified in amyloid-ß precursor protein (APP), presenilin 1 (PSEN1) and presenilin 2 (PSEN2). Additionally, common and rare variants occur within APP, PSEN1, and PSEN2 that may be risk factors, protective factors, or benign, non-pathogenic polymorphisms. Yet, to date, no single study has carefully examined the effect of all of the variants of unknown significance reported in APP, PSEN1 and PSEN2 on Aß isoform levels in vitro. In this study, we analyzed Aß isoform levels by ELISA in a cell-based system in which each reported pathogenic and risk variant in APP, PSEN1, and PSEN2 was expressed individually. In order to classify variants for which limited family history data is available, we have implemented an algorithm for determining pathogenicity using available information from multiple domains, including genetic, bioinformatic, and in vitro analyses. We identified 90 variants of unknown significance and classified 19 as likely pathogenic mutations. We also propose that five variants are possibly protective. In defining a subset of these variants as pathogenic, individuals from these families may eligible to enroll in observational studies and clinical trials.


Asunto(s)
Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Presenilina-1/genética , Presenilina-2/genética , Humanos , Mutación
5.
Biol Psychiatry ; 83(4): 300-310, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28666525

RESUMEN

Alzheimer's disease (AD) is a genetically heterogeneous neurodegenerative disorder caused by fully penetrant single gene mutations in a minority of cases, while the majority of cases are sporadic or show modest familial clustering. These cases are of late onset and likely result from the interaction of many genes and the environment. More than 30 loci have been implicated in AD by a combination of linkage, genome-wide association, and whole genome/exome sequencing. We have learned from these studies that perturbations in endolysosomal, lipid metabolism, and immune response pathways substantially contribute to sporadic AD pathogenesis. We review here current knowledge about functions of AD susceptibility genes, highlighting cells of the myeloid lineage as drivers of at least part of the genetic component in late-onset AD. Although targeted resequencing utilized for the identification of causal variants has discovered coding mutations in some AD-associated genes, a lot of risk variants lie in noncoding regions. Here we discuss the use of functional genomics approaches that integrate transcriptomic, epigenetic, and endophenotype traits with systems biology to annotate genetic variants, and to facilitate discovery of AD risk genes. Further validation in cell culture and mouse models will be necessary to establish causality for these genes. This knowledge will allow mechanism-based design of novel therapeutic interventions in AD and promises coherent implementation of treatment in a personalized manner.


Asunto(s)
Enfermedad de Alzheimer/genética , Estudio de Asociación del Genoma Completo , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Humanos
6.
Nat Neurosci ; 21(8): 1049-1060, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30038282

RESUMEN

The rapid elimination of dying neurons and nonfunctional synapses in the brain is carried out by microglia, the resident myeloid cells of the brain. Here we show that microglia clearance activity in the adult brain is regionally regulated and depends on the rate of neuronal attrition. Cerebellar, but not striatal or cortical, microglia exhibited high levels of basal clearance activity, which correlated with an elevated degree of cerebellar neuronal attrition. Exposing forebrain microglia to apoptotic cells activated gene-expression programs supporting clearance activity. We provide evidence that the polycomb repressive complex 2 (PRC2) epigenetically restricts the expression of genes that support clearance activity in striatal and cortical microglia. Loss of PRC2 leads to aberrant activation of a microglia clearance phenotype, which triggers changes in neuronal morphology and behavior. Our data highlight a key role of epigenetic mechanisms in preventing microglia-induced neuronal alterations that are frequently associated with neurodegenerative and psychiatric diseases.


Asunto(s)
Encéfalo/fisiología , Epigénesis Genética/fisiología , Microglía/fisiología , Animales , Apoptosis/genética , Muerte Celular/genética , Cerebelo/citología , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Femenino , Regulación de la Expresión Génica/genética , Activación de Macrófagos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Neostriado/citología , Neostriado/fisiología , Neostriado/ultraestructura , Neuronas/fisiología , Neuronas/ultraestructura , Complejo Represivo Polycomb 2/genética , Convulsiones/genética , Sinapsis/fisiología
7.
Stem Cell Reports ; 9(2): 600-614, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28757165

RESUMEN

Growing evidence implicates the importance of glia, particularly astrocytes, in neurological and psychiatric diseases. Here, we describe a rapid and robust method for the differentiation of highly pure populations of replicative astrocytes from human induced pluripotent stem cells (hiPSCs), via a neural progenitor cell (NPC) intermediate. We evaluated this protocol across 42 NPC lines (derived from 30 individuals). Transcriptomic analysis demonstrated that hiPSC-astrocytes from four individuals are highly similar to primary human fetal astrocytes and characteristic of a non-reactive state. hiPSC-astrocytes respond to inflammatory stimulants, display phagocytic capacity, and enhance microglial phagocytosis. hiPSC-astrocytes also possess spontaneous calcium transient activity. Our protocol is a reproducible, straightforward (single medium), and rapid (<30 days) method to generate populations of hiPSC-astrocytes that can be used for neuron-astrocyte and microglia-astrocyte co-cultures for the study of neuropsychiatric disorders.


Asunto(s)
Astrocitos/citología , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Astrocitos/metabolismo , Calcio/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Células Cultivadas , Biología Computacional/métodos , Citocinas/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Microglía/inmunología , Microglía/metabolismo , Anotación de Secuencia Molecular , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Fagocitosis , Transcriptoma
8.
Nat Neurosci ; 20(8): 1052-1061, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28628103

RESUMEN

A genome-wide survival analysis of 14,406 Alzheimer's disease (AD) cases and 25,849 controls identified eight previously reported AD risk loci and 14 novel loci associated with age at onset. Linkage disequilibrium score regression of 220 cell types implicated the regulation of myeloid gene expression in AD risk. The minor allele of rs1057233 (G), within the previously reported CELF1 AD risk locus, showed association with delayed AD onset and lower expression of SPI1 in monocytes and macrophages. SPI1 encodes PU.1, a transcription factor critical for myeloid cell development and function. AD heritability was enriched within the PU.1 cistrome, implicating a myeloid PU.1 target gene network in AD. Finally, experimentally altered PU.1 levels affected the expression of mouse orthologs of many AD risk genes and the phagocytic activity of mouse microglial cells. Our results suggest that lower SPI1 expression reduces AD risk by regulating myeloid gene expression and cell function.


Asunto(s)
Enfermedad de Alzheimer/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Haplotipos/genética , Polimorfismo de Nucleótido Simple/genética , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Alelos , Animales , Femenino , Estudio de Asociación del Genoma Completo/métodos , Humanos , Desequilibrio de Ligamiento/genética , Masculino , Ratones , Factores de Riesgo , Factores de Transcripción/genética
9.
PLoS One ; 9(1): e87014, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24466315

RESUMEN

Proteolytic processing of the amyloid precursor protein (APP) by the ß- and γ-secretases releases the amyloid-ß peptide (Aß), which deposits in senile plaques and contributes to the etiology of Alzheimer's disease (AD). The α-secretase cleaves APP in the Aß peptide sequence to generate soluble APPα (sAPPα). Upregulation of α-secretase activity through the 5-hydroxytryptamine 4 (5-HT4) receptor has been shown to reduce Aß production, amyloid plaque load and to improve cognitive impairment in transgenic mouse models of AD. Consequently, activation of 5-HT4 receptors following agonist stimulation is considered to be a therapeutic strategy for AD treatment; however, the signaling cascade involved in 5-HT4 receptor-stimulated proteolysis of APP remains to be determined. Here we used chemical and siRNA inhibition to identify the proteins which mediate 5-HT4d receptor-stimulated α-secretase activity in the SH-SY5Y human neuronal cell line. We show that G protein and Src dependent activation of phospholipase C are required for α-secretase activity, while, unexpectedly, adenylyl cyclase and cAMP are not involved. Further elucidation of the signaling pathway indicates that inositol triphosphate phosphorylation and casein kinase 2 activation is also a prerequisite for α-secretase activity. Our findings provide a novel route to explore the treatment of AD through 5-HT4 receptor-induced α-secretase activation.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Serotonina/metabolismo , Transducción de Señal/fisiología , Adenilil Ciclasas/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Arrestinas/metabolismo , Quinasa de la Caseína II/metabolismo , Línea Celular Tumoral , AMP Cíclico/metabolismo , Proteínas de Unión al GTP/metabolismo , Humanos , Inositol Polifosfato 5-Fosfatasas , Ratones , Ratones Transgénicos/metabolismo , Ratones Transgénicos/fisiología , Monoéster Fosfórico Hidrolasas/metabolismo , Proteolisis , Receptores de Serotonina 5-HT4/metabolismo , Fosfolipasas de Tipo C/metabolismo , beta-Arrestinas , Familia-src Quinasas/metabolismo
10.
Neurobiol Aging ; 34(7): 1779-89, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23474291

RESUMEN

Lowering the production and accumulation of Aß has been explored as treatment for Alzheimer's disease (AD), because Aß is postulated to play an important role in the pathogenesis of AD. 5-HT4 receptors are an interesting drug target in this regard, as their activation might stimulate α-secretase processing, which increases sAPPα and reduces Aß, at least according to the central dogma in APP processing. Here we describe a novel high-affinity 5-HT4 receptor agonist SSP-002392 that, in cultured human neuroblastoma cells, potently increases the levels of cAMP and sAPPα at 100-fold lower concentrations than the effective concentrations of prucalopride, a known selective 5-HT4 receptor agonist. Chronic administration of this compound in a hAPP/PS1 mouse model of Alzheimer's disease decreased soluble and insoluble Aß in hippocampus, but the potential mechanisms underlying these observations seem to be complex. We found no evidence for direct α-secretase stimulation in the brain in vivo, but observed decreased APP and BACE-1 expression and elevated astroglia and microglia responses. Taken together these results provide support for a potential disease-modifying aspect when stimulating central 5-HT4 receptors; however, the complexity of the phenomena warrants further research.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/biosíntesis , Péptidos beta-Amiloides/metabolismo , Presenilina-1/genética , Receptores de Serotonina 5-HT4/metabolismo , Agonistas de Receptores de Serotonina/farmacología , Agonistas de Receptores de Serotonina/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/antagonistas & inhibidores , Precursor de Proteína beta-Amiloide/biosíntesis , Precursor de Proteína beta-Amiloide/genética , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA