Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Neurodegener ; 19(1): 37, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38654375

RESUMO

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.


Assuntos
Doença de Alzheimer , Microglia , Oligonucleotídeos Antissenso , Microglia/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/genética , Humanos , Oligonucleotídeos Antissenso/farmacologia , Animais , Camundongos , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Modelos Animais de Doenças
2.
Nat Neurosci ; 27(5): 886-900, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38539015

RESUMO

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.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Microglia , Receptores Imunológicos , Transcriptoma , Humanos , Microglia/metabolismo , Microglia/patologia , Doença de Alzheimer/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/genética , Animais , Peptídeos beta-Amiloides/metabolismo , Camundongos , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Camundongos Transgênicos , Xenoenxertos , Placa Amiloide/patologia , Placa Amiloide/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia
3.
Mol Cell ; 83(22): 4106-4122.e10, 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37977120

RESUMO

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.


Assuntos
Doença de Alzheimer , Secretases da Proteína Precursora do Amiloide , Camundongos , Animais , Humanos , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Proteoma/genética , Transdução de Sinais , Proteínas de Membrana/metabolismo , Doença de Alzheimer/genética
4.
Nat Protoc ; 16(2): 1013-1033, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33424025

RESUMO

Microglia are critically involved in complex neurological disorders with a strong genetic component, such as Alzheimer's disease, Parkinson's disease and frontotemporal dementia. Although mouse microglia can recapitulate aspects of human microglia physiology, they do not fully capture the human genetic aspects of disease and do not reproduce all human cell states. Primary cultures of human microglia or microglia derived from human induced pluripotent stem cells (PSCs) are difficult to maintain in brain-relevant cell states in vitro. Here we describe MIGRATE (microglia in vitro generation refined for advanced transplantation experiments, which provides a combined in vitro differentiation and in vivo xenotransplantation protocol to study human microglia in the context of the mouse brain. This article details an accurate, step-by-step workflow that includes in vitro microglia differentiation from human PSCs, transplantation into the mouse brain and quantitative analysis of engraftment. Compared to current differentiation and xenotransplantation protocols, we present an optimized, faster and more efficient approach that yields up to 80% chimerism. To quantitatively assess engraftment efficiency by flow cytometry, access to specialized flow cytometry is required. Alternatively, the percentage of chimerism can be estimated by standard immunohistochemical analysis. The MIGRATE protocol takes ~40 d to complete, from culturing PSCs to engraftment efficiency assessment.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Microglia/citologia , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Encéfalo/fisiologia , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Microglia/metabolismo , Microglia/fisiologia , Células-Tronco Pluripotentes/citologia , Gravidez
5.
Cell Rep ; 32(13): 108189, 2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32997994

RESUMO

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.


Assuntos
Perfilação da Expressão Gênica/métodos , Microglia/fisiologia , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Humanos
6.
EMBO Mol Med ; 12(3): e10606, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-31951107

RESUMO

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.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Microglia , Proteínas tau , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Modelos Animais de Doenças , Redes Reguladoras de Genes , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Proteínas tau/metabolismo
7.
Nat Neurosci ; 22(12): 2111-2116, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31659342

RESUMO

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.


Assuntos
Doença de Alzheimer/genética , Células-Tronco Embrionárias/citologia , Microglia/metabolismo , Microglia/transplante , Transcriptoma , Peptídeos beta-Amiloides/farmacologia , Animais , Diferenciação Celular , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos
8.
Cell Rep ; 27(4): 1293-1306.e6, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31018141

RESUMO

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.


Assuntos
Envelhecimento/patologia , Doença de Alzheimer/patologia , Biomarcadores/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Microglia/patologia , Placa Amiloide/patologia , Envelhecimento/genética , Envelhecimento/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Biomarcadores/análise , Encéfalo/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Camundongos Transgênicos , Microglia/metabolismo , Placa Amiloide/genética , Placa Amiloide/metabolismo , Presenilinas/fisiologia , Caracteres Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...