Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Cell Rep ; 43(6): 114216, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38819990

RESUMEN

The amyloid plaque niche is a pivotal hallmark of Alzheimer's disease (AD). Here, we employ two high-resolution spatial transcriptomics (ST) platforms, CosMx and Spatial Enhanced Resolution Omics-sequencing (Stereo-seq), to characterize the transcriptomic alterations, cellular compositions, and signaling perturbations in the amyloid plaque niche in an AD mouse model. We discover heterogeneity in the cellular composition of plaque niches, marked by an increase in microglial accumulation. We profile the transcriptomic alterations of glial cells in the vicinity of plaques and conclude that the microglial response to plaques is consistent across different brain regions, while the astrocytic response is more heterogeneous. Meanwhile, as the microglial density of plaque niches increases, astrocytes acquire a more neurotoxic phenotype and play a key role in inducing GABAergic signaling and decreasing glutamatergic signaling in hippocampal neurons. We thus show that the accumulation of microglia around hippocampal plaques disrupts astrocytic signaling, in turn inducing an imbalance in neuronal synaptic signaling.


Asunto(s)
Enfermedad de Alzheimer , Astrocitos , Modelos Animales de Enfermedad , Microglía , Placa Amiloide , Transcriptoma , Animales , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/genética , Microglía/metabolismo , Microglía/patología , Astrocitos/metabolismo , Astrocitos/patología , Placa Amiloide/metabolismo , Placa Amiloide/patología , Transcriptoma/genética , Ratones , Hipocampo/metabolismo , Hipocampo/patología , Ratones Transgénicos , Comunicación Celular , Transducción de Señal , Neuronas/metabolismo , Neuronas/patología , Masculino
2.
Mol Neurodegener ; 19(1): 37, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38654375

RESUMEN

BACKGROUND: Microglia play important roles in maintaining brain homeostasis and neurodegeneration. The discovery of genetic variants in genes predominately or exclusively expressed in myeloid cells, such as Apolipoprotein E (APOE) and triggering receptor expressed on myeloid cells 2 (TREM2), as the strongest risk factors for Alzheimer's disease (AD) highlights the importance of microglial biology in the brain. The sequence, structure and function of several microglial proteins are poorly conserved across species, which has hampered the development of strategies aiming to modulate the expression of specific microglial genes. One way to target APOE and TREM2 is to modulate their expression using antisense oligonucleotides (ASOs). METHODS: In this study, we identified, produced, and tested novel, selective and potent ASOs for human APOE and TREM2. We used a combination of in vitro iPSC-microglia models, as well as microglial xenotransplanted mice to provide proof of activity in human microglial in vivo. RESULTS: We proved their efficacy in human iPSC microglia in vitro, as well as their pharmacological activity in vivo in a xenografted microglia model. We demonstrate ASOs targeting human microglia can modify their transcriptional profile and their response to amyloid-ß plaques in vivo in a model of AD. CONCLUSIONS: This study is the first proof-of-concept that human microglial can be modulated using ASOs in a dose-dependent manner to manipulate microglia phenotypes and response to neurodegeneration in vivo.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Oligonucleótidos Antisentido , Microglía/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Humanos , Oligonucleótidos Antisentido/farmacología , Animales , Ratones , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Células Madre Pluripotentes Inducidas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Modelos Animales de Enfermedad
3.
Nat Neurosci ; 27(5): 886-900, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38539015

RESUMEN

Microglia are central players in Alzheimer's disease pathology but analyzing microglial states in human brain samples is challenging due to genetic diversity, postmortem delay and admixture of pathologies. To circumvent these issues, here we generated 138,577 single-cell expression profiles of human stem cell-derived microglia xenotransplanted in the brain of the AppNL-G-F model of amyloid pathology and wild-type controls. Xenografted human microglia adopt a disease-associated profile similar to that seen in mouse microglia, but display a more pronounced human leukocyte antigen or HLA state, likely related to antigen presentation in response to amyloid plaques. The human microglial response also involves a pro-inflammatory cytokine/chemokine cytokine response microglia or CRM response to oligomeric Aß oligomers. Genetic deletion of TREM2 or APOE as well as APOE polymorphisms and TREM2R47H expression in the transplanted microglia modulate these responses differentially. The expression of other Alzheimer's disease risk genes is differentially regulated across the distinct cell states elicited in response to amyloid pathology. Thus, we have identified multiple transcriptomic cell states adopted by human microglia in a multipronged response to Alzheimer's disease-related pathology, which should be taken into account in translational studies.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Microglía , Transcriptoma , Animales , Humanos , Ratones , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Xenoinjertos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Placa Amiloide/patología , Placa Amiloide/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo
4.
Mol Cell ; 83(22): 4106-4122.e10, 2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-37977120

RESUMEN

γ-Secretases mediate the regulated intramembrane proteolysis (RIP) of more than 150 integral membrane proteins. We developed an unbiased γ-secretase substrate identification (G-SECSI) method to study to what extent these proteins are processed in parallel. We demonstrate here parallel processing of at least 85 membrane proteins in human microglia in steady-state cell culture conditions. Pharmacological inhibition of γ-secretase caused substantial changes of human microglial transcriptomes, including the expression of genes related to the disease-associated microglia (DAM) response described in Alzheimer disease (AD). While the overall effects of γ-secretase deficiency on transcriptomic cell states remained limited in control conditions, exposure of mouse microglia to AD-inducing amyloid plaques strongly blocked their capacity to mount this putatively protective DAM cell state. We conclude that γ-secretase serves as a critical signaling hub integrating the effects of multiple extracellular stimuli into the overall transcriptome of the cell.


Asunto(s)
Enfermedad de Alzheimer , Secretasas de la Proteína Precursora del Amiloide , Ratones , Animales , Humanos , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Proteoma/genética , Transducción de Señal , Proteínas de la Membrana/metabolismo , Enfermedad de Alzheimer/genética
5.
Science ; 381(6663): 1176-1182, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37708272

RESUMEN

Neuronal cell loss is a defining feature of Alzheimer's disease (AD), but the underlying mechanisms remain unclear. We xenografted human or mouse neurons into the brain of a mouse model of AD. Only human neurons displayed tangles, Gallyas silver staining, granulovacuolar neurodegeneration (GVD), phosphorylated tau blood biomarkers, and considerable neuronal cell loss. The long noncoding RNA MEG3 was strongly up-regulated in human neurons. This neuron-specific long noncoding RNA is also up-regulated in AD patients. MEG3 expression alone was sufficient to induce necroptosis in human neurons in vitro. Down-regulation of MEG3 and inhibition of necroptosis using pharmacological or genetic manipulation of receptor-interacting protein kinase 1 (RIPK1), RIPK3, or mixed lineage kinase domain-like protein (MLKL) rescued neuronal cell loss in xenografted human neurons. This model suggests potential therapeutic approaches for AD and reveals a human-specific vulnerability to AD.


Asunto(s)
Enfermedad de Alzheimer , Necroptosis , Neuronas , ARN Largo no Codificante , Animales , Humanos , Ratones , Enfermedad de Alzheimer/patología , Xenoinjertos , Necroptosis/genética , Neuronas/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética
6.
Nat Neurosci ; 26(6): 1021-1031, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37188873

RESUMEN

Early Alzheimer's disease (AD) is associated with hippocampal hyperactivity and decreased sleep quality. Here we show that homeostatic mechanisms transiently counteract the increased excitatory drive to CA1 neurons in AppNL-G-F mice, but that this mechanism fails in older mice. Spatial transcriptomics analysis identifies Pmch as part of the adaptive response in AppNL-G-F mice. Pmch encodes melanin-concentrating hormone (MCH), which is produced in sleep-active lateral hypothalamic neurons that project to CA1 and modulate memory. We show that MCH downregulates synaptic transmission, modulates firing rate homeostasis in hippocampal neurons and reverses the increased excitatory drive to CA1 neurons in AppNL-G-F mice. AppNL-G-F mice spend less time in rapid eye movement (REM) sleep. AppNL-G-F mice and individuals with AD show progressive changes in morphology of CA1-projecting MCH axons. Our findings identify the MCH system as vulnerable in early AD and suggest that impaired MCH-system function contributes to aberrant excitatory drive and sleep defects, which can compromise hippocampus-dependent functions.


Asunto(s)
Enfermedad de Alzheimer , Hormonas Hipotalámicas , Ratones , Animales , Enfermedad de Alzheimer/genética , Neuronas/fisiología , Hormonas Hipofisarias , Sueño , Ratones Transgénicos
7.
iScience ; 26(6): 106829, 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37250784

RESUMEN

microRNA-132 (miR-132), a known neuronal regulator, is one of the most robustly downregulated microRNAs (miRNAs) in the brain of Alzheimer's disease (AD) patients. Increasing miR-132 in AD mouse brain ameliorates amyloid and Tau pathologies, and also restores adult hippocampal neurogenesis and memory deficits. However, the functional pleiotropy of miRNAs requires in-depth analysis of the effects of miR-132 supplementation before it can be moved forward for AD therapy. We employ here miR-132 loss- and gain-of-function approaches using single-cell transcriptomics, proteomics, and in silico AGO-CLIP datasets to identify molecular pathways targeted by miR-132 in mouse hippocampus. We find that miR-132 modulation significantly affects the transition of microglia from a disease-associated to a homeostatic cell state. We confirm the regulatory role of miR-132 in shifting microglial cell states using human microglial cultures derived from induced pluripotent stem cells.

8.
Cells ; 12(6)2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36980299

RESUMEN

In malignant cancer, excessive amounts of mutant p53 often lead to its aggregation, a feature that was recently identified as druggable. Here, we describe that induction of a heat shock-related stress response mediated by Foldlin, a small-molecule tool compound, reduces the protein levels of misfolded/aggregated mutant p53, while contact mutants or wild-type p53 remain largely unaffected. Foldlin also prevented the formation of stress-induced p53 nuclear inclusion bodies. Despite our inability to identify a specific molecular target, Foldlin also reduced protein levels of aggregating SOD1 variants. Finally, by screening a library of 778 FDA-approved compounds for their ability to reduce misfolded mutant p53, we identified the proteasome inhibitor Bortezomib with similar cellular effects as Foldlin. Overall, the induction of a cellular heat shock response seems to be an effective strategy to deal with pathological protein aggregation. It remains to be seen however, how this strategy can be translated to a clinical setting.


Asunto(s)
Pliegue de Proteína , Proteína p53 Supresora de Tumor , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Inhibidores de Proteasoma/farmacología , Respuesta al Choque Térmico , Bortezomib/farmacología
9.
Cell Rep ; 40(8): 111280, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-36001964

RESUMEN

Dysfunctions of network activity and functional connectivity (FC) represent early events in Alzheimer's disease (AD), but the underlying mechanisms remain unclear. Astrocytes regulate local neuronal activity in the healthy brain, but their involvement in early network hyperactivity in AD is unknown. We show increased FC in the human cingulate cortex several years before amyloid deposition. We find the same early cingulate FC disruption and neuronal hyperactivity in AppNL-F mice. Crucially, these network disruptions are accompanied by decreased astrocyte calcium signaling. Recovery of astrocytic calcium activity normalizes neuronal hyperactivity and FC, as well as seizure susceptibility and day/night behavioral disruptions. In conclusion, we show that astrocytes mediate initial features of AD and drive clinically relevant phenotypes.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Animales , Astrocitos/metabolismo , Calcio/metabolismo , Señalización del Calcio , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Neuronas/metabolismo
10.
Nat Commun ; 12(1): 4506, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34301930

RESUMEN

Polygenic Risk Scores (PRS) for AD offer unique possibilities for reliable identification of individuals at high and low risk of AD. However, there is little agreement in the field as to what approach should be used for genetic risk score calculations, how to model the effect of APOE, what the optimal p-value threshold (pT) for SNP selection is and how to compare scores between studies and methods. We show that the best prediction accuracy is achieved with a model with two predictors (APOE and PRS excluding APOE region) with pT<0.1 for SNP selection. Prediction accuracy in a sample across different PRS approaches is similar, but individuals' scores and their associated ranking differ. We show that standardising PRS against the population mean, as opposed to the sample mean, makes the individuals' scores comparable between studies. Our work highlights the best strategies for polygenic profiling when assessing individuals for AD risk.


Asunto(s)
Enfermedad de Alzheimer/genética , Apolipoproteínas E/genética , Estudio de Asociación del Genoma Completo/métodos , Herencia Multifactorial/genética , Polimorfismo de Nucleótido Simple , Alelos , Enfermedad de Alzheimer/diagnóstico , Estudios de Casos y Controles , Frecuencia de los Genes , Genética de Población/métodos , Genética de Población/estadística & datos numéricos , Estudio de Asociación del Genoma Completo/estadística & datos numéricos , Genotipo , Humanos , Reproducibilidad de los Resultados , Medición de Riesgo/métodos , Medición de Riesgo/estadística & datos numéricos , Factores de Riesgo , Sensibilidad y Especificidad
11.
Cell Stem Cell ; 28(10): 1805-1821.e8, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34033742

RESUMEN

Neural stem cells residing in the hippocampal neurogenic niche sustain lifelong neurogenesis in the adult brain. Adult hippocampal neurogenesis (AHN) is functionally linked to mnemonic and cognitive plasticity in humans and rodents. In Alzheimer's disease (AD), the process of generating new neurons at the hippocampal neurogenic niche is impeded, yet the mechanisms involved are unknown. Here we identify miR-132, one of the most consistently downregulated microRNAs in AD, as a potent regulator of AHN, exerting cell-autonomous proneurogenic effects in adult neural stem cells and their progeny. Using distinct AD mouse models, cultured human primary and established neural stem cells, and human patient material, we demonstrate that AHN is directly affected by AD pathology. miR-132 replacement in adult mouse AD hippocampus restores AHN and relevant memory deficits. Our findings corroborate the significance of AHN in mouse models of AD and reveal the possible therapeutic potential of targeting miR-132 in neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer , MicroARNs , Enfermedad de Alzheimer/genética , Animales , Modelos Animales de Enfermedad , Hipocampo , Humanos , Trastornos de la Memoria/genética , Trastornos de la Memoria/terapia , Ratones , MicroARNs/genética , Neurogénesis
12.
Cell Rep ; 32(13): 108189, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32997994

RESUMEN

Single-nucleus RNA sequencing (snRNA-seq) is used as an alternative to single-cell RNA-seq, as it allows transcriptomic profiling of frozen tissue. However, it is unclear whether snRNA-seq is able to detect cellular state in human tissue. Indeed, snRNA-seq analyses of human brain samples have failed to detect a consistent microglial activation signature in Alzheimer's disease. Our comparison of microglia from single cells and single nuclei of four human subjects reveals that, although most genes show similar relative abundances in cells and nuclei, a small population of genes (∼1%) is depleted in nuclei compared to whole cells. This population is enriched for genes previously implicated in microglial activation, including APOE, CST3, SPP1, and CD74, comprising 18% of previously identified microglial-disease-associated genes. Given the low sensitivity of snRNA-seq to detect many activation genes, we conclude that snRNA-seq is not suited for detecting cellular activation in microglia in human disease.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Microglía/fisiología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Humanos
13.
Cell ; 182(4): 976-991.e19, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32702314

RESUMEN

Although complex inflammatory-like alterations are observed around the amyloid plaques of Alzheimer's disease (AD), little is known about the molecular changes and cellular interactions that characterize this response. We investigate here, in an AD mouse model, the transcriptional changes occurring in tissue domains in a 100-µm diameter around amyloid plaques using spatial transcriptomics. We demonstrate early alterations in a gene co-expression network enriched for myelin and oligodendrocyte genes (OLIGs), whereas a multicellular gene co-expression network of plaque-induced genes (PIGs) involving the complement system, oxidative stress, lysosomes, and inflammation is prominent in the later phase of the disease. We confirm the majority of the observed alterations at the cellular level using in situ sequencing on mouse and human brain sections. Genome-wide spatial transcriptomics analysis provides an unprecedented approach to untangle the dysregulated cellular network in the vicinity of pathogenic hallmarks of AD and other brain diseases.


Asunto(s)
Enfermedad de Alzheimer/patología , Análisis de Secuencia de ADN/métodos , Transcriptoma , Enfermedad de Alzheimer/genética , Amiloide/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Lisosomas/genética , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vaina de Mielina/genética , Vaina de Mielina/metabolismo , Estrés Oxidativo/genética
14.
EMBO Mol Med ; 12(3): e10606, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-31951107

RESUMEN

Polygenic risk scores have identified that genetic variants without genome-wide significance still add to the genetic risk of developing Alzheimer's disease (AD). Whether and how subthreshold risk loci translate into relevant disease pathways is unknown. We investigate here the involvement of AD risk variants in the transcriptional responses of two mouse models: APPswe/PS1L166P and Thy-TAU22. A unique gene expression module, highly enriched for AD risk genes, is specifically responsive to Aß but not TAU pathology. We identify in this module 7 established AD risk genes (APOE, CLU, INPP5D, CD33, PLCG2, SPI1, and FCER1G) and 11 AD GWAS genes below the genome-wide significance threshold (GPC2, TREML2, SYK, GRN, SLC2A5, SAMSN1, PYDC1, HEXB, RRBP1, LYN, and BLNK), that become significantly upregulated when exposed to Aß. Single microglia sequencing confirms that Aß, not TAU, pathology induces marked transcriptional changes in microglia, including increased proportions of activated microglia. We conclude that genetic risk of AD functionally translates into different microglia pathway responses to Aß pathology, placing AD genetic risk downstream of the amyloid pathway but upstream of TAU pathology.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Microglía , Proteínas tau , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Redes Reguladoras de Genes , Hipocampo/metabolismo , Masculino , Ratones , Ratones Transgénicos , Microglía/metabolismo , Proteínas tau/metabolismo
15.
Nat Neurosci ; 22(12): 2111-2116, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31659342

RESUMEN

Although genetics highlights the role of microglia in Alzheimer's disease, one-third of putative Alzheimer's disease risk genes lack adequate mouse orthologs. Here we successfully engraft human microglia derived from embryonic stem cells in the mouse brain. The cells recapitulate transcriptionally human primary microglia ex vivo and show expression of human-specific Alzheimer's disease risk genes. Oligomeric amyloid-ß induces a divergent response in human versus mouse microglia. This model can be used to study the role of microglia in neurological diseases.


Asunto(s)
Enfermedad de Alzheimer/genética , Células Madre Embrionarias/citología , Microglía/metabolismo , Microglía/trasplante , Transcriptoma , Péptidos beta-Amiloides/farmacología , Animales , Diferenciación Celular , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos
16.
Nat Commun ; 10(1): 2542, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31186416

RESUMEN

Somatic ribosomal protein mutations have recently been described in cancer, yet their impact on cellular transcription and translation remains poorly understood. Here, we integrate mRNA sequencing, ribosome footprinting, polysomal RNA sequencing and mass spectrometry datasets from a mouse lymphoid cell model to characterize the T-cell acute lymphoblastic leukemia (T-ALL) associated ribosomal RPL10 R98S mutation. Surprisingly, RPL10 R98S induces changes in protein levels primarily through transcriptional rather than translation efficiency changes. Phosphoserine phosphatase (PSPH), encoding a key serine biosynthesis enzyme, was the only gene with elevated transcription and translation leading to protein overexpression. PSPH upregulation is a general phenomenon in T-ALL patient samples, associated with elevated serine and glycine levels in xenograft mice. Reduction of PSPH expression suppresses proliferation of T-ALL cell lines and their capacity to expand in mice. We identify ribosomal mutation driven induction of serine biosynthesis and provide evidence supporting dependence of T-ALL cells on PSPH.


Asunto(s)
Glicina/metabolismo , Mutación , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Serina/metabolismo , Animales , Línea Celular , Perfilación de la Expresión Génica , Ratones , Monoéster Fosfórico Hidrolasas , Polirribosomas/genética , Polirribosomas/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/genética , Proteína Ribosómica L10 , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Ribosomas/metabolismo , Análisis de Secuencia de ARN
17.
Cell Rep ; 27(4): 1293-1306.e6, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31018141

RESUMEN

Gene expression profiles of more than 10,000 individual microglial cells isolated from cortex and hippocampus of male and female AppNL-G-F mice over time demonstrate that progressive amyloid-ß accumulation accelerates two main activated microglia states that are also present during normal aging. Activated response microglia (ARMs) are composed of specialized subgroups overexpressing MHC type II and putative tissue repair genes (Dkk2, Gpnmb, and Spp1) and are strongly enriched with Alzheimer's disease (AD) risk genes. Microglia from female mice progress faster in this activation trajectory. Similar activated states are also found in a second AD model and in human brain. Apoe, the major genetic risk factor for AD, regulates the ARMs but not the interferon response microglia (IRMs). Thus, the ARMs response is the converging point for aging, sex, and genetic AD risk factors.


Asunto(s)
Envejecimiento/patología , Enfermedad de Alzheimer/patología , Biomarcadores/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Microglía/patología , Placa Amiloide/patología , Envejecimiento/genética , Envejecimiento/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/fisiología , Animales , Biomarcadores/análisis , Encéfalo/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Ratones Transgénicos , Microglía/metabolismo , Placa Amiloide/genética , Placa Amiloide/metabolismo , Presenilinas/fisiología , Caracteres Sexuales
18.
Alzheimers Dement ; 15(3): 453-464, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30442540

RESUMEN

INTRODUCTION: Murine microglia expressing the Alzheimer's disease-linked TREM2R47H mutation display variable decrease in phagocytosis, while impaired phagocytosis is reported following loss of TREM2. However, no data exist on TREM2+/R47H human microglia. Therefore, we created human pluripotent stem cell (hPSC) monocytes and transdifferentiated microglia-like cells (tMGs) to examine the effect of the TREM2+/R47H mutation and loss of TREM2 on phagocytosis. METHODS: We generated isogenic TREM2+/R47H, TREM2+/-, and TREM2-/- hPSCs using CRISPR/Cas9. Following differentiation to monocytes and tMGs, we studied the uptake of Escherichia coli fragments and analyzed amyloid plaque clearance from cryosections of APP/PS1+/- mouse brains. RESULTS: We demonstrated that tMGs resemble cultured human microglia. TREM2+/- and TREM2-/- hPSC monocytes and tMGs phagocytosed significantly less E. coli fragments and cleared less amyloid plaques than wild-type hPSC progeny, with no difference for TREM2+/R47H progeny. DISCUSSION: In vitro phagocytosis of hPSC monocytes and tMGs was not affected by the TREM2+/R47H mutation but was significantly impaired in TREM2+/- and TREM2-/- progeny.


Asunto(s)
Glicoproteínas de Membrana/deficiencia , Microglía/metabolismo , Monocitos/metabolismo , Placa Amiloide/metabolismo , Receptores Inmunológicos/deficiencia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo , Sistemas CRISPR-Cas , Células Cultivadas , Escherichia coli , Glicoproteínas de Membrana/genética , Ratones Transgénicos , Fagocitosis , Células Madre Pluripotentes , Presenilina-1/genética , Presenilina-1/metabolismo , Receptores Inmunológicos/genética
20.
Mol Neurodegener ; 13(1): 54, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30314521

RESUMEN

BACKGROUND: Despite diverging levels of amyloid-ß (Aß) and TAU pathology, different mouse models, as well as sporadic AD patients show predictable patterns of episodic memory loss. MicroRNA (miRNA) deregulation is well established in AD brain but it is unclear whether Aß or TAU pathology drives those alterations and whether miRNA changes contribute to cognitive decline. METHODS: miRNAseq was performed on cognitively intact (4 months) and impaired (10 months) male APPtg (APPswe/PS1L166P) and TAUtg (THY-Tau22) mice and their wild-type littermates (APPwt and TAUwt). We analyzed the hippocampi of 12 mice per experimental group (n = 96 in total), and employed a 2-way linear model to extract differentially expressed miRNAs. Results were confirmed by qPCR in a separate cohort of 4 M and 10 M APPtg and APPwt mice (n = 7-9 per group) and in human sporadic AD and non-demented control brain. Fluorescent in situ hybridization identified their cellular expression. Functional annotation of predicted targets was performed using GO enrichment. Behavior of wild-type mice was assessed after intracerebroventricular infusion of miRNA mimics. RESULTS: Six miRNAs (miR-10a-5p, miR-142a-5p, miR-146a-5p, miR-155-5p, miR-211-5p, miR-455-5p) are commonly upregulated between APPtg and TAUtg mice, and four of these (miR-142a-5p, miR-146a-5p, miR-155-5p and miR-455-5p) are altered in AD patients. All 6 miRNAs are strongly enriched in neurons. Upregulating these miRNAs in wild-type mice is however not causing AD-related cognitive disturbances. CONCLUSION: Diverging AD-related neuropathologies induce common disturbances in the expression of neuronal miRNAs. 4 of these miRNAs are also upregulated in AD patients. Therefore these 4 miRNAs (miR-142a-5p, miR-146a-5p, miR-155-5p and miR-455-5p) appear part of a core pathological process in AD patients and APPtg and TAUtg mice. They are however not causing cognitive disturbances in wild-type mice. As some of these miRNA target AD relevant proteins, they may be, in contrast, part of a protective response in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , MicroARNs/genética , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Animales , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Ratones Transgénicos , Neuronas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...