Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 16313, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34381098

RESUMO

The first night effect (FNE) is a type of sleep disturbance caused by an unfamiliar environment, which leads to difficulty falling asleep and reduced sleep duration. Previously, we reported that Lactobacillus fermentum PS150 (PS150) improves sleep conditions in a pentobarbital-induced sleep mouse model. In this study, we aimed to evaluate the effect of PS150 on the FNE in mice. Briefly, mice were implanted with electrodes and orally administered PS150 for four weeks, and then the FNE was induced by cage changing. Analysis of polysomnographic signals revealed that intervention with PS150 restored non-rapid eye movement (NREM) sleep length under the FNE. Compared to diphenhydramine, a commonly used sleep aid, PS150 had no unwanted side effects, such as rapid eye movement (REM) sleep deprivation and fragmented sleep. Moreover, temporal analysis revealed that PS150 efficiently reduced both sleep latency and time spent restoring normal levels of REM sleep. Taken together, these results suggest that PS150 efficiently ameliorates sleep disturbance caused by the FNE. Additionally, V3-V4 16S rRNA sequencing revealed significant increases in Erysipelotrichia, Actinobacteria, and Coriobacteriia in fecal specimens of the PS150-treated group, indicating that PS150 induces gut microbiota remodeling.


Assuntos
Limosilactobacillus fermentum/fisiologia , Sono REM/fisiologia , Animais , Modelos Animais de Doenças , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pentobarbital/farmacologia , Polissonografia/métodos , RNA Ribossômico 16S/genética , Privação do Sono/induzido quimicamente , Privação do Sono/microbiologia , Privação do Sono/fisiopatologia , Distúrbios do Início e da Manutenção do Sono/induzido quimicamente , Distúrbios do Início e da Manutenção do Sono/microbiologia , Distúrbios do Início e da Manutenção do Sono/fisiopatologia , Transtornos do Sono-Vigília/induzido quimicamente , Transtornos do Sono-Vigília/microbiologia , Transtornos do Sono-Vigília/fisiopatologia , Sono REM/efeitos dos fármacos
2.
Physiol Genomics ; 52(7): 280-292, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32567509

RESUMO

The gut microbiota, via the production of metabolites entering the circulation, plays a role in blood pressure regulation. Blood pressure is also affected by the characteristics of sleep. To date, no studies have examined relationships among the gut microbiota/metabolites, blood pressure, and sleep. We hypothesized that fragmented sleep is associated with elevated mean arterial pressure, an altered and dysbiotic gut microbial community, and changes in fecal metabolites. In our model system, rats were randomized to 8 h of sleep fragmentation during the rest phase (light phase) or were undisturbed (controls) for 28 consecutive days. Rats underwent sleep and blood pressure recordings, and fecal samples were analyzed during: baseline (days -4 to -1), early sleep fragmentation (days 0-3), midsleep fragmentation (days 6-13), late sleep fragmentation (days 20-27), and recovery/rest (days 28-34). Less sleep per hour during the sleep fragmentation period was associated with increased mean arterial pressure. Analyses of gut microbial communities and metabolites revealed that putative short chain fatty acid-producing bacteria were differentially abundant between control and intervention animals during mid-/late sleep fragmentation and recovery. Midsleep fragmentation was also characterized by lower alpha diversity, lower Firmicutes:Bacteroidetes ratio, and higher Proteobacteria in intervention rats. Elevated putative succinate-producing bacteria and acetate-producing bacteria were associated with lower and higher mean arterial pressure, respectively, and untargeted metabolomics analysis demonstrates that certain fecal metabolites are significantly correlated with blood pressure. These data reveal associations between sleep fragmentation, mean arterial pressure, and the gut microbiome/fecal metabolome and provide insight to links between disrupted sleep and cardiovascular pathology.


Assuntos
Pressão Sanguínea , Disbiose/microbiologia , Fezes/microbiologia , Microbioma Gastrointestinal , Metaboloma , Privação do Sono/metabolismo , Privação do Sono/microbiologia , Acetatos/metabolismo , Animais , Bactérias/genética , Bactérias/metabolismo , Ácidos Graxos Voláteis/metabolismo , Masculino , Metabolômica , RNA Ribossômico 16S , Ratos , Ratos Endogâmicos WKY , Ácido Succínico/metabolismo
3.
J Microbiol ; 58(7): 588-597, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32424577

RESUMO

Our study demonstrated that sleep deprivation resulted in homeostasis disorder of colon. Our study goes deeper into the positive effects of melatonin on small intestinal microbiota disorder caused by sleep deprivation. We successfully established a multiplatform 72 h sleep deprivation mouse model with or without melatonin supplementation, and analyzed the change of small intestinal microbiota using high-throughput sequencing of the 16S rRNA. We found melatonin supplementation suppressed the decrease of plasma melatonin level in sleep deprivation mice. Meanwhile, melatonin supplementation improved significantly the reduction in OTU numbers and the diversity and richness of jejunal microbiota and the abundance of Bacteroidaeae and Prevotellaceae, as well as an increase in the Firmicutes-to-Bacteroidetes ratio and the content of Moraxellaceae and Aeromonadaceae in the jejunum of sleep deprived-mice. Moreover, melatonin supplementation reversed the change of metabolic pathway in sleep deprived-mice, including metabolism, signal transduction mechanisms and transcription etc, which were related to intestinal health. Furthermore, melatonin supplementation inverted the sleep deprivation-induced a decline of anti-inflammatory cytokines (IL-22) and an increase of the ROS and proinflammatory cytokines (IL-17) in jejunum. These findings suggested that melatonin, similar to a probiotics agent, can reverse sleep deprivation-induced small intestinal microbiota disorder by suppressing oxidative stress and inflammation response.


Assuntos
Antioxidantes/farmacologia , Disbiose/tratamento farmacológico , Microbioma Gastrointestinal/efeitos dos fármacos , Jejuno/microbiologia , Melatonina/farmacologia , Privação do Sono/microbiologia , Aeromonadaceae/efeitos dos fármacos , Aeromonadaceae/isolamento & purificação , Animais , Bacteroidaceae/efeitos dos fármacos , Bacteroidaceae/isolamento & purificação , Firmicutes/efeitos dos fármacos , Firmicutes/isolamento & purificação , Inflamação , Interleucina-17/análise , Interleucinas/análise , Masculino , Camundongos , Camundongos Endogâmicos ICR , Moraxellaceae/efeitos dos fármacos , Moraxellaceae/isolamento & purificação , Estresse Oxidativo/efeitos dos fármacos , Prevotella/efeitos dos fármacos , Prevotella/isolamento & purificação , RNA Ribossômico 16S/genética , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética
4.
PLoS One ; 15(2): e0229001, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32078624

RESUMO

It has been established in recent years that the gut microbiome plays a role in health and disease, potentially via alterations in metabolites that influence host physiology. Although sleep disruption and gut dysbiosis have been associated with many of the same diseases, studies investigating the gut microbiome in the context of sleep disruption have yielded inconsistent results, and have not assessed the fecal metabolome. We exposed mice to five days of sleep disruption followed by four days of ad libitum recovery sleep, and assessed the fecal microbiome and fecal metabolome at multiple timepoints using 16S rRNA gene amplicons and untargeted LC-MS/MS mass spectrometry. We found global shifts in both the microbiome and metabolome in the sleep-disrupted group on the second day of recovery sleep, when most sleep parameters had recovered to baseline levels. We observed an increase in the Firmicutes:Bacteroidetes ratio, along with decreases in the genus Lactobacillus, phylum Actinobacteria, and genus Bifidobacterium in sleep-disrupted mice compared to control mice. The latter two taxa remained low at the fourth day post-sleep disruption. We also identified multiple classes of fecal metabolites that were differentially abundant in sleep-disrupted mice, some of which are physiologically relevant and commonly influenced by the microbiome. This included bile acids, and inference of microbial functional gene content suggested reduced levels of the microbial bile salt hydrolase gene in sleep-disrupted mice. Overall, this study adds to the evidence base linking disrupted sleep to the gut microbiome and expands it to the fecal metabolome, identifying sleep disruption-sensitive bacterial taxa and classes of metabolites that may serve as therapeutic targets to improve health after poor sleep.


Assuntos
Bactérias , Fezes/microbiologia , Microbioma Gastrointestinal , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , Privação do Sono/microbiologia , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Masculino , Camundongos
5.
Gut Microbes ; 11(4): 706-720, 2020 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-31924109

RESUMO

Sleep is a fundamental biological process, that when repeatedly disrupted, can result in severe health consequences. Recent studies suggest that both sleep fragmentation (SF) and dysbiosis of the gut microbiome can lead to metabolic disorders, though the underlying mechanisms are largely unclear. To better understand the consequences of SF, we investigated the effects of acute (6 days) and chronic (6 weeks) SF on rats by examining taxonomic profiles of microbiota in the distal ileum, cecum and proximal colon, as well as assessing structural and functional integrity of the gastrointestinal barrier. We further assayed the impact of SF on a host function by evaluating inflammation and immune response. Both acute and chronic SF induced microbial dysbiosis, more dramatically in the distal ileum (compared to other two regions studied), as noted by significant perturbations in alpha- and beta-diversity; though, specific microbial populations were significantly altered throughout each of the three regions. Furthermore, chronic SF resulted in increased crypt depth in the distal ileum and an increase in the number of villi lining both the cecum and proximal colon. Additional changes were noted with chronic SF, including: decreased microbial adhesion and penetration in the distal ileum and cecum, elevation in serum levels of the cytokine KC/GRO, and depressed levels of corticotropin. Importantly, our data show that perturbations to microbial ecology and intestinal morphology intensify in response to prolonged SF and these changes are habitat specific. Together, these results reveal consequences to gut microbiota homeostasis and host response following acute and chronic SF in rats.


Assuntos
Ceco/patologia , Colo/patologia , Microbioma Gastrointestinal , Íleo/patologia , Privação do Sono , Animais , Aderência Bacteriana , Fenômenos Fisiológicos Bacterianos , Ceco/microbiologia , Colo/microbiologia , Citocinas/análise , Endotoxinas/análise , Sistema Hipotálamo-Hipofisário/fisiologia , Íleo/microbiologia , Filogenia , Ratos , Ratos Sprague-Dawley , Privação do Sono/microbiologia , Privação do Sono/patologia , Privação do Sono/fisiopatologia
6.
J Pineal Res ; 67(1): e12574, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30929267

RESUMO

Intestinal diseases caused by sleep deprivation (SD) are severe public health threats worldwide. This study focuses on the effect of melatonin on intestinal mucosal injury and microbiota dysbiosis in sleep-deprived mice. Mice subjected to SD had significantly elevated norepinephrine levels and decreased melatonin content in plasma. Consistent with the decrease in melatonin levels, we observed a decrease of antioxidant ability, down-regulation of anti-inflammatory cytokines and up-regulation of pro-inflammatory cytokines in sleep-deprived mice, which resulted in colonic mucosal injury, including a reduced number of goblet cells, proliferating cell nuclear antigen-positive cells, expression of MUC2 and tight junction proteins and elevated expression of ATG5, Beclin1, p-P65 and p-IκB. High-throughput pyrosequencing of 16S rRNA demonstrated that the diversity and richness of the colonic microbiota were decreased in sleep-deprived mice, especially in probiotics, including Akkermansia, Bacteroides and Faecalibacterium. However, the pathogen Aeromonas was markedly increased. By contrast, supplementation with 20 and 40 mg/kg melatonin reversed these SD-induced changes and improved the mucosal injury and dysbiosis of the microbiota in the colon. Our results suggest that the effect of SD on intestinal barrier dysfunction might be an outcome of melatonin suppression rather than a loss of sleep per se. SD-induced intestinal barrier dysfunction involved the suppression of melatonin production and activation of the NF-κB pathway by oxidative stress.


Assuntos
Colo , Microbioma Gastrointestinal/imunologia , Enteropatias , Mucosa Intestinal , Melatonina/imunologia , Privação do Sono , Animais , Colo/imunologia , Colo/microbiologia , Enteropatias/imunologia , Enteropatias/microbiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Probióticos , Privação do Sono/imunologia , Privação do Sono/microbiologia
7.
Physiol Res ; 67(Suppl 3): S409-S420, 2018 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-30484668

RESUMO

Obesity is linked to a wide range of serious illnesses. In addition to the important impact on the health of the individual, obesity also has a substantial impact on the economy. Disruption of physiological day-night cycles could contribute to the increased incidence of obesity. According to the American National Sleep Federation, the percentage of the people who reported a sleep duration of six hours or less increased from 12 to 37 % over ten years. Insufficient sleep leads not only to an increase of the total calorie intake but changes the meal preference in favor of palatable foods and meals with high carbohydrate content. A decrease of leptin and increase of ghrelin levels caused by sleep deficiency can also play a role. In addition to the higher caloric intake, the timing of food consumption should be taken into account. The same meal eaten during the night versus the day is associated with increased postprandial glucose and triglyceride levels. The gut microbiome has also been recently understood as an endocrine system, with links between the gut microbiome and circadian rhythm changes possibly influencing increased obesity.


Assuntos
Ritmo Circadiano/fisiologia , Microbioma Gastrointestinal/fisiologia , Obesidade/microbiologia , Ingestão de Energia/fisiologia , Humanos , Obesidade/complicações , Obesidade/metabolismo , Período Pós-Prandial/fisiologia , Privação do Sono/complicações , Privação do Sono/metabolismo , Privação do Sono/microbiologia
8.
Proc Natl Acad Sci U S A ; 114(8): E1564-E1571, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28179566

RESUMO

Insufficient sleep increasingly characterizes modern society, contributing to a host of serious medical problems. Loss of sleep is associated with metabolic diseases such as obesity and diabetes, cardiovascular disorders, and neurological and cognitive impairments. Shifts in gut microbiome composition have also been associated with the same pathologies; therefore, we hypothesized that sleep restriction may perturb the gut microbiome to contribute to a disease state. In this study, we examined the fecal microbiome by using a cross-species approach in both rat and human studies of sleep restriction. We used DNA from hypervariable regions (V1-V2) of 16S bacteria rRNA to define operational taxonomic units (OTUs) of the microbiome. Although the OTU richness of the microbiome is decreased by sleep restriction in rats, major microbial populations are not altered. Only a single OTU, TM7-3a, was found to increase with sleep restriction of rats. In the human microbiome, we find no overt changes in the richness or composition induced by sleep restriction. Together, these results suggest that the microbiome is largely resistant to changes during sleep restriction.


Assuntos
Cognição/fisiologia , Disbiose/fisiopatologia , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/microbiologia , Doenças Metabólicas/fisiopatologia , Privação do Sono/fisiopatologia , Adulto , Animais , DNA Bacteriano/isolamento & purificação , Disbiose/microbiologia , Fezes/microbiologia , Feminino , Trato Gastrointestinal/fisiopatologia , Genes de RNAr , Voluntários Saudáveis , Humanos , Masculino , Doenças Metabólicas/microbiologia , Pessoa de Meia-Idade , RNA Ribossômico 16S/genética , Ratos , Ratos Sprague-Dawley , Privação do Sono/microbiologia
9.
Sci Rep ; 6: 35405, 2016 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-27739530

RESUMO

Chronic sleep fragmentation (SF) commonly occurs in human populations, and although it does not involve circadian shifts or sleep deprivation, it markedly alters feeding behaviors ultimately promoting obesity and insulin resistance. These symptoms are known to be related to the host gut microbiota. Mice were exposed to SF for 4 weeks and then allowed to recover for 2 weeks. Taxonomic profiles of fecal microbiota were obtained prospectively, and conventionalization experiments were performed in germ-free mice. Adipose tissue insulin sensitivity and inflammation, as well as circulating measures of inflammation, were assayed. Effect of fecal water on colonic epithelial permeability was also examined. Chronic SF-induced increased food intake and reversible gut microbiota changes characterized by the preferential growth of highly fermentative members of Lachnospiraceae and Ruminococcaceae and a decrease of Lactobacillaceae families. These lead to systemic and visceral white adipose tissue inflammation in addition to altered insulin sensitivity in mice, most likely via enhanced colonic epithelium barrier disruption. Conventionalization of germ-free mice with SF-derived microbiota confirmed these findings. Thus, SF-induced metabolic alterations may be mediated, in part, by concurrent changes in gut microbiota, thereby opening the way for gut microbiome-targeted therapeutics aimed at reducing the major end-organ morbidities of chronic SF.


Assuntos
Tecido Adiposo/metabolismo , Microbioma Gastrointestinal , Resistência à Insulina , Privação do Sono/microbiologia , Animais , Insulina/sangue , Interleucinas/sangue , Lactobacillaceae/isolamento & purificação , Leptina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Privação do Sono/sangue , Privação do Sono/metabolismo
11.
Am J Physiol Regul Integr Comp Physiol ; 278(4): R905-16, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10749778

RESUMO

Profound sleep disruption in humans is generally believed to cause health impairments. Through comparative research, specific physical effects and underlying mechanisms altered by sleep deprivation are being elucidated. Studies of sleep-deprived animals previously have shown a progressive, chronic negative energy balance and gradual deterioration of health, which culminate in fatal bloodstream infection without an infectious focus. The present study investigated the conditions antecedent to advanced morbidity in sleep-deprived rats by determining the time course and distribution of live microorganisms in body tissues that are normally sterile. The tissues cultured for microbial growth included the blood, four major organs, six regional lymph nodes, the intestine, and the skin. The principal finding was early infection of the mesenteric lymph nodes by bacteria presumably translocated from the intestine and bacterial migration to and transient infection of extraintestinal sites. Presence of pathogenic microorganisms and their toxins in tissues constitutes a septic burden and chronic antigenic challenge for the host. Bacterial translocation and pathogenic sequelae provide mechanisms by which sleep deprivation appears to adversely affect health.


Assuntos
Infecções Bacterianas/imunologia , Infecções Bacterianas/fisiopatologia , Imunidade/fisiologia , Privação do Sono/imunologia , Privação do Sono/microbiologia , Animais , Bactérias Aeróbias/isolamento & purificação , Bactérias Anaeróbias/isolamento & purificação , Técnicas Bacteriológicas , Peso Corporal , Doença Crônica , Ingestão de Alimentos , Bactérias Gram-Negativas/isolamento & purificação , Intestinos/microbiologia , Rim/microbiologia , Fígado/microbiologia , Linfonodos/microbiologia , Masculino , Neuroimunomodulação/imunologia , Ratos , Ratos Sprague-Dawley , Pele/microbiologia , Baço/microbiologia , Cauda
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...