Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
Tipo de estudo
Intervalo de ano de publicação
1.
J Neuroinflammation ; 20(1): 3, 2023 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-36593462

RESUMO

BACKGROUND: Astrocytes are crucial for maintaining brain homeostasis and synaptic function, but are also tightly connected to the pathogenesis of Alzheimer's disease (AD). Our previous data demonstrate that astrocytes ingest large amounts of aggregated amyloid-beta (Aß), but then store, rather than degrade the ingested material, which leads to severe cellular stress. However, the involvement of pathological astrocytes in AD-related synaptic dysfunction remains to be elucidated. METHODS: In this study, we aimed to investigate how intracellular deposits of Aß in astrocytes affect their interplay with neurons, focusing on neuronal function and viability. For this purpose, human induced pluripotent stem cell (hiPSC)-derived astrocytes were exposed to sonicated Αß42 fibrils. The direct and indirect effects of the Αß-exposed astrocytes on hiPSC-derived neurons were analyzed by performing astrocyte-neuron co-cultures as well as additions of conditioned media or extracellular vesicles to pure neuronal cultures. RESULTS: Electrophysiological recordings revealed significantly decreased frequency of excitatory post-synaptic currents in neurons co-cultured with Aß-exposed astrocytes, while conditioned media from Aß-exposed astrocytes had the opposite effect and resulted in hyperactivation of the synapses. Clearly, factors secreted from control, but not from Aß-exposed astrocytes, benefited the wellbeing of neuronal cultures. Moreover, reactive astrocytes with Aß deposits led to an elevated clearance of dead cells in the co-cultures. CONCLUSIONS: Taken together, our results demonstrate that inclusions of aggregated Aß affect the reactive state of the astrocytes, as well as their ability to support neuronal function.


Assuntos
Doença de Alzheimer , Células-Tronco Pluripotentes Induzidas , Humanos , Astrócitos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Meios de Cultivo Condicionados/farmacologia , Células Cultivadas , Peptídeos beta-Amiloides/farmacologia , Peptídeos beta-Amiloides/metabolismo , Neurônios/metabolismo , Doença de Alzheimer/patologia
2.
J Neuroinflammation ; 18(1): 124, 2021 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-34082772

RESUMO

BACKGROUND: Alzheimer's disease (AD) and Parkinson's disease (PD) are characterized by brain accumulation of aggregated amyloid-beta (Aß) and alpha-synuclein (αSYN), respectively. In order to develop effective therapies, it is crucial to understand how the Aß/αSYN aggregates can be cleared. Compelling data indicate that neuroinflammatory cells, including astrocytes and microglia, play a central role in the pathogenesis of AD and PD. However, how the interplay between the two cell types affects their clearing capacity and consequently the disease progression remains unclear. METHODS: The aim of the present study was to investigate in which way glial crosstalk influences αSYN and Aß pathology, focusing on accumulation and degradation. For this purpose, human-induced pluripotent cell (hiPSC)-derived astrocytes and microglia were exposed to sonicated fibrils of αSYN or Aß and analyzed over time. The capacity of the two cell types to clear extracellular and intracellular protein aggregates when either cultured separately or in co-culture was studied using immunocytochemistry and ELISA. Moreover, the capacity of cells to interact with and process protein aggregates was tracked using time-lapse microscopy and a customized "close-culture" chamber, in which the apical surfaces of astrocyte and microglia monocultures were separated by a <1 mm space. RESULTS: Our data show that intracellular deposits of αSYN and Aß are significantly reduced in co-cultures of astrocytes and microglia, compared to monocultures of either cell type. Analysis of conditioned medium and imaging data from the "close-culture" chamber experiments indicate that astrocytes secrete a high proportion of their internalized protein aggregates, while microglia do not. Moreover, co-cultured astrocytes and microglia are in constant contact with each other via tunneling nanotubes and other membrane structures. Notably, our live cell imaging data demonstrate that microglia, when attached to the cell membrane of an astrocyte, can attract and clear intracellular protein deposits from the astrocyte. CONCLUSIONS: Taken together, our data demonstrate the importance of astrocyte and microglia interactions in Aß/αSYN clearance, highlighting the relevance of glial cellular crosstalk in the progression of AD- and PD-related brain pathology.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Astrócitos/patologia , Microglia/metabolismo , Microglia/patologia , Agregados Proteicos , Agregação Patológica de Proteínas , alfa-Sinucleína/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Encéfalo/metabolismo , Estruturas da Membrana Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Humanos , Células-Tronco Pluripotentes Induzidas , Microscopia Confocal , Nanotubos , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Proteólise
3.
Acta Neuropathol Commun ; 12(1): 34, 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38409026

RESUMO

Accumulating evidence highlights the involvement of astrocytes in Alzheimer's disease (AD) progression. We have previously demonstrated that human iPSC-derived astrocytes ingest and modify synthetic tau fibrils in a way that enhances their seeding efficiency. However, synthetic tau fibrils differ significantly from in vivo formed fibrils. To mimic the situation in the brain, we here analyzed astrocytes' processing of human brain-derived tau fibrils and its consequences for cellular physiology. Tau fibrils were extracted from both AD and control brains, aiming to examine any potential differences in astrocyte response depending on the origin of fibrils. Our results show that human astrocytes internalize, but fail to degrade, both AD and control tau fibrils. Instead, pathogenic, seeding capable tau proteoforms are spread to surrounding cells via tunneling nanotubes and exocytosis. Notably, accumulation of AD tau fibrils induces a stronger reactive state in astrocytes, compared to control fibrils, evident by the augmented expression of vimentin and GFAP, as well as by an increased secretion of the pro-inflammatory cytokines IL-8 and MCP-1. Moreover, conditioned media from astrocytes with AD tau fibril deposits induce synapse and metabolic impairment in human iPSC-derived neurons. Taken together, our data suggest that the accumulation of brain-derived AD tau fibrils induces a more robust inflammatory and neurotoxic phenotype in human astrocytes, accentuating the nature of tau fibrils as an important contributing factor to inflammation and neurodegeneration in AD.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/patologia , Astrócitos/metabolismo , Proteínas tau/metabolismo , Encéfalo/patologia , Neurônios/metabolismo , Inflamação/metabolismo
4.
Acta Neuropathol Commun ; 11(1): 97, 2023 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-37330529

RESUMO

Tau deposits in astrocytes are frequently found in Alzheimer's disease (AD) and other tauopathies. Since astrocytes do not express tau, the inclusions have been suggested to be of neuronal origin. However, the mechanisms behind their appearance and their relevance for disease progression remain unknown. Here we demonstrate, using a battery of experimental techniques that human astrocytes serve as an intermediator, promoting cell-to-cell spreading of pathological tau. Human astrocytes engulf and process, but fail to fully degrade dead neurons with tau pathology, as well as synthetic tau fibrils and tau aggregates isolated from AD brain tissue. Instead, the pathogenic tau is spread to nearby cells via secretion and tunneling nanotube mediated transfer. By performing co-culture experiments we could show that tau-containing astrocytes induce tau pathology in healthy human neurons directly. Furthermore, our results from a FRET based seeding assay, demonstrated that the tau proteoforms secreted by astrocytes have an exceptional seeding capacity, compared to the original tau species engulfed by the cells. Taken together, our study establishes a central role for astrocytes in mediating tau pathology, which could be of relevance for identifying novel treatment targets for AD and other tauopathies.


Assuntos
Doença de Alzheimer , Tauopatias , Humanos , Proteínas tau/metabolismo , Astrócitos/metabolismo , Tauopatias/patologia , Doença de Alzheimer/patologia , Neurônios/metabolismo , Encéfalo/patologia
5.
Environ Sci Pollut Res Int ; 27(20): 25261-25270, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32347502

RESUMO

Bisphenol A diglycidyl ether (BADGE), a derivative of bisphenol A (BPA), is widely used in the manufacture of epoxy resins as well as a coating on food containers. Recent studies have demonstrated the adverse effects of BADGE on reproduction and development in rodents and amphibians, but how BADGE affects biological activity is not understood. To gain a better understanding of the biological effects of BADGE exposure during development, we used the model organism Drosophila melanogaster and performed whole transcriptome sequencing. Interestingly, when Drosophila are raised on food containing BADGE, genes having significantly increased transcript numbers are enriched for those involved in regulating cell proliferation, including DNA replication and cell cycle control. Furthermore, raising larvae on BADGE-containing food induces hemocyte (blood cell) over-proliferation. This effect can be stimulated with even lower concentrations of BADGE if the hemocytes are already primed for cell proliferation by the expression of dominant active Ras GTPase. We conclude that chronic exposure to the xenobiotic BADGE throughout development can induce cell proliferation.


Assuntos
Drosophila , Poluentes Ambientais , Animais , Compostos Benzidrílicos , Proliferação de Células , Drosophila melanogaster , Compostos de Epóxi
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA