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.
J Mol Med (Berl) ; 95(1): 29-39, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27858116

RESUMO

The microbiome located within a given host and its organs forms a holobiont, an intimate functional entity with evolutionarily designed interactions to support nutritional intake and reproduction. Thus, all organs in a holobiont respond to changes within the microbiome. The development and function of the central nervous system and its homeostatic mechanisms are no exception and are also subject to regulation by the gut microbiome. In order for the holobiont to function effectively, the microbiome and host must communicate. The aryl hydrocarbon receptor is an evolutionarily conserved receptor recognizing environmental compounds, including a number of ligands produced directly and indirectly by the microbiome. This review focuses on the microbiome-gut-brain axis in regard to the aryl hydrocarbon receptor signaling pathway and its impact on underlying mechanisms in neurodegeneration.


Assuntos
Sistema Nervoso Central/fisiologia , Interações Hospedeiro-Patógeno , Microbiota , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais , Animais , Microbioma Gastrointestinal , Trato Gastrointestinal , Humanos , Ligantes , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurogênese
3.
Sci Rep ; 6: 20127, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26879573

RESUMO

The liver is a key organ of metabolic homeostasis with functions that oscillate in response to food intake. Although liver and gut microbiome crosstalk has been reported, microbiome-mediated effects on peripheral circadian clocks and their output genes are less well known. Here, we report that germ-free (GF) mice display altered daily oscillation of clock gene expression with a concomitant change in the expression of clock output regulators. Mice exposed to microbes typically exhibit characterized activities of nuclear receptors, some of which (PPARα, LXRß) regulate specific liver gene expression networks, but these activities are profoundly changed in GF mice. These alterations in microbiome-sensitive gene expression patterns are associated with daily alterations in lipid, glucose, and xenobiotic metabolism, protein turnover, and redox balance, as revealed by hepatic metabolome analyses. Moreover, at the systemic level, daily changes in the abundance of biomarkers such as HDL cholesterol, free fatty acids, FGF21, bilirubin, and lactate depend on the microbiome. Altogether, our results indicate that the microbiome is required for integration of liver clock oscillations that tune output activators and their effectors, thereby regulating metabolic gene expression for optimal liver function.


Assuntos
Relógios Circadianos/genética , Fígado/metabolismo , Microbiota , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais , Animais , Biomarcadores , Feminino , Microbioma Gastrointestinal , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Gluconeogênese/genética , Inativação Metabólica/genética , Masculino , Camundongos , Especificidade de Órgãos , Receptores Citoplasmáticos e Nucleares/genética , Transcriptoma
4.
NPJ Biofilms Microbiomes ; 2: 16014, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28721249

RESUMO

The ligand-induced transcription factor, aryl hydrocarbon receptor (AhR) is known for its capacity to tune adaptive immunity and xenobiotic metabolism-biological properties subject to regulation by the indigenous microbiome. The objective of this study was to probe the postulated microbiome-AhR crosstalk and whether such an axis could influence metabolic homeostasis of the host. Utilising a systems-biology approach combining in-depth 1H-NMR-based metabonomics (plasma, liver and skeletal muscle) with microbiome profiling (small intestine, colon and faeces) of AhR knockout (AhR-/-) and wild-type (AhR+/+) mice, we assessed AhR function in host metabolism. Microbiome metabolites such as short-chain fatty acids were found to regulate AhR and its target genes in liver and intestine. The AhR signalling pathway, in turn, was able to influence microbiome composition in the small intestine as evident from microbiota profiling of the AhR+/+ and AhR-/- mice fed with diet enriched with a specific AhR ligand or diet depleted of any known AhR ligands. The AhR-/- mice also displayed increased levels of corticosterol and alanine in serum. In addition, activation of gluconeogenic genes in the AhR-/- mice was indicative of on-going metabolic stress. Reduced levels of ketone bodies and reduced expression of genes involved in fatty acid metabolism in the liver further underscored this observation. Interestingly, exposing AhR-/- mice to a high-fat diet showed resilience to glucose intolerance. Our data suggest the existence of a bidirectional AhR-microbiome axis, which influences host metabolic pathways.

5.
Sci Transl Med ; 6(263): 263ra158, 2014 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-25411471

RESUMO

Pivotal to brain development and function is an intact blood-brain barrier (BBB), which acts as a gatekeeper to control the passage and exchange of molecules and nutrients between the circulatory system and the brain parenchyma. The BBB also ensures homeostasis of the central nervous system (CNS). We report that germ-free mice, beginning with intrauterine life, displayed increased BBB permeability compared to pathogen-free mice with a normal gut flora. The increased BBB permeability was maintained in germ-free mice after birth and during adulthood and was associated with reduced expression of the tight junction proteins occludin and claudin-5, which are known to regulate barrier function in endothelial tissues. Exposure of germ-free adult mice to a pathogen-free gut microbiota decreased BBB permeability and up-regulated the expression of tight junction proteins. Our results suggest that gut microbiota-BBB communication is initiated during gestation and propagated throughout life.


Assuntos
Barreira Hematoencefálica , Intestinos/microbiologia , Microbiota , Animais , Feminino , Camundongos , Permeabilidade , Gravidez , Junções Íntimas/metabolismo
6.
PLoS Pathog ; 10(1): e1003887, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24465207

RESUMO

To be able to colonize its host, invading Salmonella enterica serovar Typhimurium must disrupt and severely affect host-microbiome homeostasis. Here we report that S. Typhimurium induces acute infectious colitis by inhibiting peroxisome proliferator-activated receptor gamma (PPARγ) expression in intestinal epithelial cells. Interestingly, this PPARγ down-regulation by S. Typhimurium is independent of TLR-4 signaling but triggers a marked elevation of host innate immune response genes, including that encoding the antimicrobial peptide lipocalin-2 (Lcn2). Accumulation of Lcn2 stabilizes the metalloproteinase MMP-9 via extracellular binding, which further aggravates the colitis. Remarkably, when exposed to S. Typhimurium, Lcn2-null mice exhibited a drastic reduction of the colitis and remained protected even at later stages of infection. Our data suggest a mechanism in which S. Typhimurium hijacks the control of host immune response genes such as those encoding PPARγ and Lcn2 to acquire residence in a host, which by evolution has established a symbiotic relation with its microbiome community to prevent pathogen invasion.


Assuntos
Proteínas de Fase Aguda/imunologia , Colite/imunologia , Evasão da Resposta Imune , Lipocalinas/imunologia , Proteínas Oncogênicas/imunologia , PPAR gama/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Doença Aguda , Proteínas de Fase Aguda/genética , Animais , Linhagem Celular , Colite/genética , Colite/microbiologia , Colite/patologia , Humanos , Lipocalina-2 , Lipocalinas/genética , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/imunologia , Camundongos , Camundongos Knockout , Proteínas Oncogênicas/genética , PPAR gama/genética , Infecções por Salmonella/genética , Infecções por Salmonella/patologia , Salmonella typhimurium/patogenicidade , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia
7.
Am J Physiol Gastrointest Liver Physiol ; 304(11): G1025-37, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23518684

RESUMO

Short-chain fatty acids (SCFAs), such as butyrate and propionate, are metabolic products of carbohydrate fermentation by the microbiota and constitute the main source of energy for host colonocytes. SCFAs are also important for gastrointestinal health, immunity, and host metabolism. Intestinally produced angiopoietin-like protein 4 (ANGPTL4) is a secreted protein with metabolism-altering properties and may offer a route by which microbiota can regulate host metabolism. Peroxisome proliferator-activated receptor (PPAR)-γ has previously been shown to be involved in microbiota-induced expression of intestinal ANGPTL4, but the role of bacterial metabolites in this process has remained elusive. Here, we show that the SCFA butyrate regulates intestinal ANGPTL4 expression in a PPAR-γ-independent manner. Although PPAR-γ is not required for butyrate-driven intestinal ANGPTL4 expression, costimulating with PPAR-γ ligands and SCFAs leads to additive increases in ANGPTL4 levels. We suggest that PPAR-γ and butyrate rely on two separate regulatory sites, a PPAR-responsive element downstream the transcription start site and a butyrate-responsive element(s) within the promoter region, 0.5 kb upstream of the transcription start site. Furthermore, butyrate gavage and colonization with Clostridium tyrobutyricum, a SCFA producer, can independently induce expression of intestinal ANGPTL4 in germ-free mice. Thus, oral administration of SCFA or use of SCFA-producing bacteria may be additional routes to maintain intestinal ANGPTL4 levels for preventive nutrition or therapeutic purposes.


Assuntos
Angiopoietinas/metabolismo , Butiratos/farmacologia , Hipoglicemiantes/farmacologia , Mucosa Intestinal/metabolismo , Tiazolidinedionas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/genética , Animais , Células CACO-2 , Clostridium tyrobutyricum , Enterócitos/metabolismo , Vida Livre de Germes , Células HCT116 , Células HT29 , Humanos , Mucosa Intestinal/microbiologia , Metagenoma , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/agonistas , PPAR gama/metabolismo , Elementos de Resposta , Rosiglitazona , Sítio de Iniciação de Transcrição
8.
Artigo em Inglês | MEDLINE | ID: mdl-22368769

RESUMO

The gut microbiota consists of trillions of prokaryotes that reside in the intestinal mucosa. This long-established commensalism indicates that these microbes are an integral part of the eukaryotic host. Recent research findings have implicated the dynamics of microbial function in setting thresholds for many physiological parameters. Conversely, it has been convincingly argued that dysbiosis, representing microbial imbalance, may be an important underlying factor that contributes to a variety of diseases, inside and outside the gut. This review discusses the latest findings, including enterotype classification, changes brought on by dysbiosis, gut inflammation, and metabolic mediators in an attempt to underscore the importance of the gut microbiota for human health. A cautiously optimistic idea is taking hold, invoking the gut microbiota as a medium to track, target and treat a plethora of diseases.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...