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
Intervalo de ano de publicação
1.
Nature ; 595(7867): 409-414, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34194038

RESUMO

Social interactions among animals mediate essential behaviours, including mating, nurturing, and defence1,2. The gut microbiota contribute to social activity in mice3,4, but the gut-brain connections that regulate this complex behaviour and its underlying neural basis are unclear5,6. Here we show that the microbiome modulates neuronal activity in specific brain regions of male mice to regulate canonical stress responses and social behaviours. Social deviation in germ-free and antibiotic-treated mice is associated with elevated levels of the stress hormone corticosterone, which is primarily produced by activation of the hypothalamus-pituitary-adrenal (HPA) axis. Adrenalectomy, antagonism of glucocorticoid receptors, or pharmacological inhibition of corticosterone synthesis effectively corrects social deficits following microbiome depletion. Genetic ablation of glucocorticoid receptors in specific brain regions or chemogenetic inactivation of neurons in the paraventricular nucleus of the hypothalamus that produce corticotrophin-releasing hormone (CRH) reverse social impairments in antibiotic-treated mice. Conversely, specific activation of CRH-expressing neurons in the paraventricular nucleus induces social deficits in mice with a normal microbiome. Via microbiome profiling and in vivo selection, we identify a bacterial species, Enterococcus faecalis, that promotes social activity and reduces corticosterone levels in mice following social stress. These studies suggest that specific gut bacteria can restrain the activation of the HPA axis, and show that the microbiome can affect social behaviours through discrete neuronal circuits that mediate stress responses in the brain.


Assuntos
Encéfalo/citologia , Encéfalo/fisiologia , Microbioma Gastrointestinal/fisiologia , Neurônios/metabolismo , Comportamento Social , Estresse Psicológico , Animais , Corticosterona/sangue , Hormônio Liberador da Corticotropina/metabolismo , Enterococcus faecalis/metabolismo , Vida Livre de Germes , Glucocorticoides/metabolismo , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais
2.
Dev Biol ; 447(2): 137-146, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30664880

RESUMO

Neural crest cells have broad migratory and differentiative ability that differs according to their axial level of origin. However, their transient nature has limited understanding of their stem cell and self-renewal properties. While an in vitro culture method has made it possible to maintain cranial neural crest cells as self-renewing multipotent crestospheres (Kerosuo et al., 2015), these same conditions failed to preserve trunk neural crest in a stem-like state. Here we optimize culture conditions for maintenance of avian trunk crestospheres, comprised of both neural crest stem and progenitor cells. Our trunk-derived crestospheres are multipotent and display self-renewal capacity over several weeks. Trunk crestospheres display elevated expression of neural crest cell markers as compared to those characteristic of ventrolateral neural tube or mesodermal fates. Moreover, trunk crestospheres express increased levels of trunk neural crest-enriched markers as compared to cranial crestospheres. Finally, we use lentiviral transduction as a tool to manipulate gene expression in trunk crestospheres. Taken together, this method enables long-term in vitro maintenance and manipulation of multipotent trunk neural crest cells in a premigratory stem or early progenitor state. Trunk crestospheres are a valuable resource for probing mechanisms underlying neural crest stemness and lineage decisions as well as accompanying diseases.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Multipotentes/metabolismo , Crista Neural/embriologia , Células-Tronco Neurais/metabolismo , Animais , Embrião de Galinha , Galinhas , Células-Tronco Multipotentes/citologia , Crista Neural/citologia , Células-Tronco Neurais/citologia
3.
Elife ; 112022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36346385

RESUMO

Parkinson's disease (PD) is a movement disorder characterized by neuroinflammation, α-synuclein pathology, and neurodegeneration. Most cases of PD are non-hereditary, suggesting a strong role for environmental factors, and it has been speculated that disease may originate in peripheral tissues such as the gastrointestinal (GI) tract before affecting the brain. The gut microbiome is altered in PD and may impact motor and GI symptoms as indicated by animal studies, although mechanisms of gut-brain interactions remain incompletely defined. Intestinal bacteria ferment dietary fibers into short-chain fatty acids, with fecal levels of these molecules differing between PD and healthy controls and in mouse models. Among other effects, dietary microbial metabolites can modulate activation of microglia, brain-resident immune cells implicated in PD. We therefore investigated whether a fiber-rich diet influences microglial function in α-synuclein overexpressing (ASO) mice, a preclinical model with PD-like symptoms and pathology. Feeding a prebiotic high-fiber diet attenuates motor deficits and reduces α-synuclein aggregation in the substantia nigra of mice. Concomitantly, the gut microbiome of ASO mice adopts a profile correlated with health upon prebiotic treatment, which also reduces microglial activation. Single-cell RNA-seq analysis of microglia from the substantia nigra and striatum uncovers increased pro-inflammatory signaling and reduced homeostatic responses in ASO mice compared to wild-type counterparts on standard diets. However, prebiotic feeding reverses pathogenic microglial states in ASO mice and promotes expansion of protective disease-associated macrophage (DAM) subsets of microglia. Notably, depletion of microglia using a CSF1R inhibitor eliminates the beneficial effects of prebiotics by restoring motor deficits to ASO mice despite feeding a prebiotic diet. These studies uncover a novel microglia-dependent interaction between diet and motor symptoms in mice, findings that may have implications for neuroinflammation and PD.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Animais , Camundongos , alfa-Sinucleína/metabolismo , Microglia/metabolismo , Prebióticos , Substância Negra , Modelos Animais de Doenças , Dieta , Camundongos Endogâmicos C57BL
4.
J Exp Med ; 216(1): 41-59, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30385457

RESUMO

Microglia, the resident immune cells in the brain, are essential for modulating neurogenesis, influencing synaptic remodeling, and regulating neuroinflammation by surveying the brain microenvironment. Microglial dysfunction has been implicated in the onset and progression of several neurodevelopmental and neurodegenerative diseases; however, the multitude of factors and signals influencing microglial activity have not been fully elucidated. Microglia not only respond to local signals within the brain but also receive input from the periphery, including the gastrointestinal (GI) tract. Recent preclinical findings suggest that the gut microbiome plays a pivotal role in regulating microglial maturation and function, and altered microbial community composition has been reported in neurological disorders with known microglial involvement in humans. Collectively, these findings suggest that bidirectional crosstalk between the gut and the brain may influence disease pathogenesis. Herein, we discuss recent studies showing a role for the gut microbiome in modulating microglial development and function in homeostatic and disease conditions and highlight possible future research to develop novel microbial treatments for disorders of the brain.


Assuntos
Encéfalo/metabolismo , Microbioma Gastrointestinal , Intestinos/microbiologia , Microglia/metabolismo , Doenças Neurodegenerativas , Transtornos do Neurodesenvolvimento , Animais , Encéfalo/patologia , Humanos , Intestinos/patologia , Microglia/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/microbiologia , Doenças Neurodegenerativas/patologia , Transtornos do Neurodesenvolvimento/metabolismo , Transtornos do Neurodesenvolvimento/microbiologia , Transtornos do Neurodesenvolvimento/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA