Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(43): 11524-11529, 2017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-29073081

RESUMEN

Variants in the gene encoding the triggering receptor expressed on myeloid cells 2 (TREM2) were recently found to increase the risk for developing Alzheimer's disease (AD). In the brain, TREM2 is predominately expressed on microglia, and its association with AD adds to increasing evidence implicating a role for the innate immune system in AD initiation and progression. Thus far, studies have found TREM2 is protective in the response to amyloid pathology while variants leading to a loss of TREM2 function impair microglial signaling and are deleterious. However, the potential role of TREM2 in the context of tau pathology has not yet been characterized. In this study, we crossed Trem2+/+ (T2+/+) and Trem2-/- (T2-/-) mice to the PS19 human tau transgenic line (PS) to investigate whether loss of TREM2 function affected tau pathology, the microglial response to tau pathology, or neurodegeneration. Strikingly, by 9 mo of age, T2-/-PS mice exhibited significantly less brain atrophy as quantified by ventricular enlargement and preserved cortical volume in the entorhinal and piriform regions compared with T2+/+PS mice. However, no TREM2-dependent differences were observed for phosphorylated tau staining or insoluble tau levels. Rather, T2-/-PS mice exhibited significantly reduced microgliosis in the hippocampus and piriform cortex compared with T2+/+PS mice. Gene expression analyses and immunostaining revealed microglial activation was significantly attenuated in T2-/-PS mice, and there were lower levels of inflammatory cytokines and astrogliosis. These unexpected findings suggest that impairing microglial TREM2 signaling reduces neuroinflammation and is protective against neurodegeneration in the setting of pure tauopathy.


Asunto(s)
Inflamación/genética , Glicoproteínas de Membrana/metabolismo , Enfermedades Neurodegenerativas/genética , Receptores Inmunológicos/metabolismo , Tauopatías , Animales , Regulación de la Expresión Génica/fisiología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores Inmunológicos/genética
2.
Neuron ; 112(3): 384-403.e8, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-37995685

RESUMEN

Apolipoprotein E (APOE) is a strong genetic risk factor for late-onset Alzheimer's disease (LOAD). APOE4 increases and APOE2 decreases risk relative to APOE3. In the P301S mouse model of tauopathy, ApoE4 increases tau pathology and neurodegeneration when compared with ApoE3 or the absence of ApoE. However, the role of ApoE isoforms and lipid metabolism in contributing to tau-mediated degeneration is unknown. We demonstrate that in P301S tau mice, ApoE4 strongly promotes glial lipid accumulation and perturbations in cholesterol metabolism and lysosomal function. Increasing lipid efflux in glia via an LXR agonist or Abca1 overexpression strongly attenuates tau pathology and neurodegeneration in P301S/ApoE4 mice. We also demonstrate reductions in reactive astrocytes and microglia, as well as changes in cholesterol biosynthesis and metabolism in glia of tauopathy mice in response to LXR activation. These data suggest that promoting efflux of glial lipids may serve as a therapeutic approach to ameliorate tau and ApoE4-linked neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Ratones , Animales , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteína E3/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Tauopatías/tratamiento farmacológico , Tauopatías/genética , Colesterol , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Ratones Transgénicos
3.
Sci Transl Med ; 15(693): eade6285, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-37099634

RESUMEN

Sleep loss is associated with cognitive decline in the aging population and is a risk factor for Alzheimer's disease (AD). Considering the crucial role of immunomodulating genes such as that encoding the triggering receptor expressed on myeloid cells type 2 (TREM2) in removing pathogenic amyloid-ß (Aß) plaques and regulating neurodegeneration in the brain, our aim was to investigate whether and how sleep loss influences microglial function in mice. We chronically sleep-deprived wild-type mice and the 5xFAD mouse model of cerebral amyloidosis, expressing either the humanized TREM2 common variant, the loss-of-function R47H AD-associated risk variant, or without TREM2 expression. Sleep deprivation not only enhanced TREM2-dependent Aß plaque deposition compared with 5xFAD mice with normal sleeping patterns but also induced microglial reactivity that was independent of the presence of parenchymal Aß plaques. We investigated lysosomal morphology using transmission electron microscopy and found abnormalities particularly in mice without Aß plaques and also observed lysosomal maturation impairments in a TREM2-dependent manner in both microglia and neurons, suggesting that changes in sleep modified neuro-immune cross-talk. Unbiased transcriptome and proteome profiling provided mechanistic insights into functional pathways triggered by sleep deprivation that were unique to TREM2 and Aß pathology and that converged on metabolic dyshomeostasis. Our findings highlight that sleep deprivation directly affects microglial reactivity, for which TREM2 is required, by altering the metabolic ability to cope with the energy demands of prolonged wakefulness, leading to further Aß deposition, and underlines the importance of sleep modulation as a promising future therapeutic approach.


Asunto(s)
Enfermedad de Alzheimer , Amiloidosis , Ratones , Animales , Microglía/metabolismo , Privación de Sueño/complicaciones , Privación de Sueño/metabolismo , Privación de Sueño/patología , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Placa Amiloide/patología , Modelos Animales de Enfermedad , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/metabolismo
4.
J Exp Med ; 216(11): 2546-2561, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31601677

RESUMEN

Chronic activation of brain innate immunity is a prominent feature of Alzheimer's disease (AD) and primary tauopathies. However, to what degree innate immunity contributes to neurodegeneration as compared with pathological protein-induced neurotoxicity, and the requirement of a particular glial cell type in neurodegeneration, are still unclear. Here we demonstrate that microglia-mediated damage, rather than pathological tau-induced direct neurotoxicity, is the leading force driving neurodegeneration in a tauopathy mouse model. Importantly, the progression of ptau pathology is also driven by microglia. In addition, we found that APOE, the strongest genetic risk factor for AD, regulates neurodegeneration predominantly by modulating microglial activation, although a minor role of apoE in regulating ptau and insoluble tau formation independent of its immunomodulatory function was also identified. Our results suggest that therapeutic strategies targeting microglia may represent an effective approach to prevent disease progression in the setting of tauopathy.


Asunto(s)
Apolipoproteínas E/inmunología , Modelos Animales de Enfermedad , Microglía/inmunología , Enfermedades Neurodegenerativas/inmunología , Tauopatías/inmunología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Aminopiridinas/administración & dosificación , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Suplementos Dietéticos , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microglía/citología , Microglía/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Pirroles/administración & dosificación , Tauopatías/genética , Tauopatías/metabolismo , Proteínas tau/genética , Proteínas tau/inmunología , Proteínas tau/metabolismo
5.
J Exp Med ; 214(5): 1227-1238, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28416651

RESUMEN

Tauopathies are characterized by the progressive accumulation of hyperphosphorylated, aggregated forms of tau. Our laboratory has previously demonstrated that passive immunization with an anti-tau antibody, HJ8.5, decreased accumulation of pathological tau in a human P301S tau-expressing transgenic (P301S-tg) mouse model of frontotemporal dementia/tauopathy. To investigate whether the Fc domain of HJ8.5 is required for the therapeutic effect, we engineered single-chain variable fragments (scFvs) derived from HJ8.5 with variable linker lengths, all specific to human tau. Based on different binding properties, we selected two anti-tau scFvs and tested their efficacy in vivo by adeno-associated virus-mediated gene transfer to the brain of P301S-tg mice. The scFvs significantly reduced levels of hyperphosphorylated, aggregated tau in brain tissue of P301S-tg mice, associated with a decrease in detergent-soluble tau species. Interestingly, these mice showed substantial levels of scFvs in the cerebrospinal fluid without significant effects on total extracellular tau levels. Therefore, our study provides a novel strategy for anti-tau immunotherapeutics that potentially limits a detrimental proinflammatory response.


Asunto(s)
Anticuerpos de Cadena Única/inmunología , Tauopatías/inmunología , Proteínas tau/inmunología , Animales , Encéfalo/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Técnicas de Transferencia de Gen , Hipocampo/metabolismo , Masculino , Ratones , Ratones Transgénicos , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/fisiología , Tauopatías/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA