Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 52(1): 117-125, 2023 Feb 25.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-37283125

RESUMO

OBJECTIVES: To establish a mouse visceral obesity model, and to investigate the effect of animal sex on this model. METHODS: Thirty-two 4-week-old BALB/c mice were randomly divided into female control group, female high-fat group, male control group and male high-fat group with 8 mice in each group.The control groups were given ordinary diet, and the high-fat groups were given high-fat diet. After 12 weeks of feeding, body weight, visceral fat, fasting blood glucose, glucose tolerance, blood lipid and metabolism-related hormone levels were measured, and the composition of gut microbiota of mice was analyzed by 16S rRNA sequencing. RESULTS: The high fat diet resulted in a significant increase of body weight and visceral fat content in male mice; the pathological results showed significantly increased fat area, accumulation of liver fat droplets, increased total cholesterol, fasting blood glucose, oral glucose tolerance and serum insulin levels (all P<0.05), as well as significant insulin resistance (P<0.01). However, the above changes were not significant in female mice. Compared with the control groups, there was an increase in the relative abundance of obesity-related gut microbiota in the model groups (such as Blautia), and the microbiota structure changed significantly, while the changes were less obvious in female mice. CONCLUSIONS: A visceral obesity mouse model has been stably established by feeding high-fat diet in BALB/c male mice, showing visceral fat accumulation, metabolic dysfunction and gut microbiota changes; while female mice are not sensitive in this obesity model.


Assuntos
Dieta Hiperlipídica , Obesidade Abdominal , Animais , Feminino , Masculino , Camundongos , Glicemia/metabolismo , Peso Corporal , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , RNA Ribossômico 16S
2.
Neurobiol Dis ; 170: 105757, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35588989

RESUMO

Gut microbiota depletion may result in cognitive impairment and emotional disorder. This study aimed to determine the possible association between host gut microbiota, cognitive function, and emotion in various life stages and its related underlying mechanisms. Seventy-five neonatal mice were randomly divided into five groups (n = 15 per group). Mice in the vehicle group were administered distilled water from birth to death, and those in the last four groups were administered antibiotic cocktail from birth to death, from birth to postnatal day (PND) 21 (infancy), from PND 21 to 56 (adolescence), and from PND 57 to 84 (adulthood), respectively. Antibiotic exposure consistently altered the gut microbiota composition and decreased the diversity of gut microbiota. Proteobacteria were the predominant bacteria instead of Firmicutes and Bacteroidetes after antibiotic exposure in different life stages. Long-term and infant gut microbiota depletion resulted in anxiety- and depression-like behaviors, memory impairments, and increased expression of γ-aminobutyric acid type A receptor α1 of adult mice. Long-term antibiotic exposure also significantly decreased serum interleukin (IL)-1ß, IL-10, and corticosterone of adult mice. Gut microbiota depletion in adolescence resulted in anxiety-like behaviors, short-term memory decline, decreased serum interferon-γ (IFN-γ), mRNA expression of 5-hydroxytryptamine receptor 1A, and neuropeptide Y receptor Y2 in the prefrontal cortex of adult mice. Antibiotic exposure in adulthood damaged short-term memory and decreased serum IL-10, IFN-γ, and increased γ-aminobutyric acid type B receptor 1 mRNA expression of adult mice. These results suggest that antibiotic-induced gut microbiota depletion in the long term and infancy resulted in the most severe cognitive and emotional disorders followed by depletion in adolescence and adulthood. These results also suggest that gut microbes could influence host cognitive function and emotion in a life stage-dependent manner by affecting the function of the immune system, hypothalamic-pituitary-adrenal axis, and the expression of neurochemicals in the brain.


Assuntos
Disfunção Cognitiva , Microbioma Gastrointestinal , Animais , Antibacterianos/farmacologia , Comportamento Animal/fisiologia , Disfunção Cognitiva/induzido quimicamente , Microbioma Gastrointestinal/fisiologia , Sistema Hipotálamo-Hipofisário , Interleucina-10 , Camundongos , Sistema Hipófise-Suprarrenal , RNA Mensageiro , Ácido gama-Aminobutírico
3.
Wei Sheng Yan Jiu ; 51(4): 624-644, 2022 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-36047269

RESUMO

OBJECTIVE: To explore the protective effect and mechanism of Bifidobacterium bifidum TMC3115 of improving the gut microbiota disorder caused by antibiotic exposurein early life, and the possible protection of inflammatory bowel disease in adulthood in mice. METHODS: 80 newborn mice were randomly divided into 3 groups, a blank control group(n=40), a ceftriaxone exposure group(n=20), a Bifidobacterium bifidum TMC3115 intervention group(n=20). After birth, they were respectively treated with saline, ceftriaxone(100 mg/kg), and ceftriaxone(100 mg/kg) + TMC3115(1×10~9CFU/d) for 3 weeks. After 3 weeks, half of each group was randomly sacrificed, and the rest were normally fed to 6 weeks. At 6 weeks, the blank control group was randomly divided into a negative control group(n=10) and a colitis model group(n=10). The negative control group drunk pure water freely, and the other three groups were added 3% DSS to the drinking water for 4 days to induce colitis. At 6 weeks and 4 days, the remaining mice were sacrificed. The weight change, spleen coefficient, gut microbiota analysis based on second-generation sequencing and serum tumor necrosis factor-α(TNF-α), interleukin-6(IL-6), and interleukin-10(IL-10)levels of the mice at 3 weeks and after DSS intervention were recorded. In addition, the colon length and inflammation pathology score of the mice after DSS intervention were also measured. RESULTS: At 3 weeks, compared with the control, antibiotic exposure in the early life inhibited the weight gain and reduced the diversity and uniformity of the gut microbiota of the mice(P<0.05). The intervention of TMC3115 under antibiotic exposure during this period increased the relative abundance of Bifidobacterium in the intestines(P<0.05), and the effect still existed after DSS stimulation in adulthood, laying the foundation for TMC3115 to exert long-term benefits. After DSS stimulation in adulthood, mice showed significant weight gain inhibition, colon length shorteningand inflammation pathology scoreincrease compared with the negative control(P<0.05), showed the inflammatory bowel disease(IBD)model was successfully constructed. The relative abundance of beneficial bacteria such as Lactobacillus in the Bifidobacterium bifidum TMC3115 intervention group increased compared with the ceftriaxone exposure group(P<0.05), while the relative abundance of harmful bacteria such as Staphylococcus, Clostridium, and Desulfovibrio decreased(P<0.05). Furthermore, the mice exposed to antibiotic in early life produced a stronger immune response, but the mice which received TMC3115 intervention at the same time had a significant decrease in serum TNF-α and IL-6 levels and increase in IL-10 level compared with the mice which only interfered with antibiotics(P<0.05). CONCLUSION: Antibiotic exposure in early life is a negative factor for long-term inflammatory bowel disease, and TMC3115 has preventive significance for long-term inflammatory bowel disease under the background of antibiotic exposure. The mechanism of TMC3115 may be to adjust the gut microbiota and balance the immune system.


Assuntos
Bifidobacterium bifidum , Colite , Doenças Inflamatórias Intestinais , Animais , Antibacterianos , Bifidobacterium bifidum/fisiologia , Ceftriaxona , Colite/induzido quimicamente , Colite/microbiologia , Colo/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Inflamação , Doenças Inflamatórias Intestinais/patologia , Interleucina-10 , Interleucina-6 , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa , Aumento de Peso
4.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 53(5): 834-841, 2022 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-36224686

RESUMO

Objective: To investigate the effects of using Bifidobacterium bifidum TMC3115 in early life on intestinal microbiota and immune functions and the long-term impact on inflammatory bowel disease. Methods: Fourteen pregnant BALB/c mice were purchased and 84 newborn BALB/c mice were subsequently obtained. Then, the newborn mice were randomly assigned to a normal saline (NS) group and a TMC3115 group, given via oral gavage normal saline and TMC3115, respectively, at a daily volume of 0.2 mL for each mouse. About 42 mice were assigned to each group. The gavage was stopped after 3 weeks. At this point, half of the mice in each group were sacrificed, and then the remaining mice in each group were randomly divided into NS-water group, NS-DSS group, TMC3115-water group, and TMC3115-DSS group, with about 10 mice in each group. The mice were given regular feed until the end of week 6 when they were given 3% dextran sulphate sodium (DSS) ad libitum for 4 days to establish the enteritis model, while the non-modeling groups were given pure water ad libitum. The experiment ended after 6 weeks and 4 days. The weekly body mass changes of the mice were documented. The intestinal tissue at the end of the experiment and the fecal samples, spleen and serum of the mice at 3 weeks and at the end of the experiment were collected to determine the pathology scores of colonic inflammation, the composition of fecal gut microbiota, spleen organ index and the mass concentration of serum cytokines. Results: 1) At the end of the experiment, the inflammatory pathology score was significantly lower in the TMC3115-DSS group compared with that of the Saline-DSS group ( P<0.05), with less disruption of colonic crypt structures and other structures, less inflammatory infiltration, and more intact epithelial structures. 2) At 3 weeks, in comparison with those of the NS group, the relative abundance of Bifidobacteriumwas significantly higher in the feces of the TMC3115 ( P<0.05), the relative abundance of both Enterococcusand Staphylococcuswas lower ( P<0.05), the splenic organ index was significantly higher ( P<0.05), and interleukin (IL)-10 was significantly decreased ( P<0.05), while there was no significant change in IL-6 or TNF-α ( P>0.05). At the end of the experiment, in comparison with those of the NS-DSS group that undergone DSS induction, the TMC3115-DSS group had reduced relative abundance of Staphylococcus, Staphylococcus tumefaciens and Escherichia/ Shigellain the feces ( P<0.05), while the splenic organ index was significantly higher ( P<0.05), and there were no significant changes in IL-6 or TNF-α ( P>0.05). Conclusion: The use of TMC3115 in early life promotes the construction of gut microbiota in neonatal mice, thereby producing a long-term effect that alleviates colitis in mice, but the mechanisms involved are still not fully understood.


Assuntos
Bifidobacterium bifidum , Colite , Microbioma Gastrointestinal , Doenças Inflamatórias Intestinais , Animais , Colite/microbiologia , Colo , Citocinas , Sulfato de Dextrana/farmacologia , Modelos Animais de Doenças , Interleucina-6 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Solução Salina/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Água/farmacologia
5.
Water Sci Technol ; 75(12): 2829-2840, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28659523

RESUMO

Given that the common medium in existing green roofs is a single layer composed of organic and inorganic substrates, seven pilot-scale dual-substrate-layer extensive green roofs (G1-G7), which include nutrition and adsorption substrate layers, were constructed in this study. The effectiveness of porous inert substrates (activated charcoal, zeolite, pumice, lava, vermiculite and expanded perlite) used as the adsorption substrate for stormwater retention was investigated. A single-substrate-layer green roof (G8) was built for comparison with G1-G7. Despite the larger total rainfall depth (mm) of six types of simulated rains (43.2, 54.6, 76.2, 87.0, 85.2 and 86.4, respectively), the total percent retention of G1-G7 varied between 14% and 82% with an average of 43%, exhibiting better runoff-retaining capacity than G8 based on the maximum potential rainfall storage depth per unit height of adsorption substrate. Regression analysis showed that there was a logarithmic relationship between cumulative rainfall depth with non-zero runoff and stormwater retention for G1-G4 and a linear relationship for G5-G8. To enhance the water retention capacity and extend the service life of dual-substrate-layer extensive green roofs, the mixture of activated charcoal and/or pumice with expanded perlite and/or vermiculite is more suitable as the adsorption substrate than the mixture containing lava and/or zeolite.


Assuntos
Conservação dos Recursos Naturais/métodos , Movimentos da Água , Arquitetura de Instituições de Saúde , Hidrologia , Porosidade , Chuva , Eliminação de Resíduos Líquidos
6.
Water Sci Technol ; 68(12): 2691-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24355859

RESUMO

The objectives of this study are to investigate the impact of different factors on the nutrient pollutant concentrations in green roof runoff and to provide reference data for the engineering design of dual substrate layer green roofs. The data were collected from eight different trays under three kinds of artificial rains. The results showed that except for total phosphorus, dual substrate layer green roofs behaved as a sink for most of the nutrient pollutants (significant at p < 0.05), and the first-flush effect did not occur during the 27 simulated rain events. The results also revealed that the concentration of these nutrient pollutants in the runoff strongly depended on the features of the nutrient substrates used in the green roof and the depth of the adsorption substrates. Compared with the influence of the substrates, the influence of the plant density and drainage systems was small.


Assuntos
Conservação dos Recursos Naturais/métodos , Arquitetura de Instituições de Saúde , Chuva , Sedum/crescimento & desenvolvimento , Poluentes Químicos da Água/análise , Análise da Demanda Biológica de Oxigênio , China , Monitoramento Ambiental , Modelos Teóricos , Nitrogênio/análise , Fósforo/análise , Urbanização , Movimentos da Água
7.
Front Microbiol ; 13: 916824, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35935215

RESUMO

Inflammatory bowel disease (IBD) is a chronic intestinal disease characterized by microbiota disturbance and intestinal mucosal damage. The current study aimed to investigate the preventive effects of Bifidobacterium bifidum BD-1 (BD-1) against long-term IBD and possible mechanism by which it alters the gut microbiota, immune response, and mucosal barrier. Our study found that early treatment of BD-1 + Ceftri (ceftriaxone followed by BD-1) and BD-1 confers a certain protective effect against the occurrence of long-term Dextran sulfate sodium-induced colitis, which manifests as a decrease in inflammation scores and MPO activity levels, as well as a relatively intact intestinal epithelial structure. Moreover, compared to BD-1, Ceftri, and NS, early treatment with BD-1 + Ceftri promoted greater expression levels of mucosal barrier-related proteins [KI67, MUC2, ZO-1, secretory immunoglobulin A (slgA), Clauding-1, and Occludin], better local immune responses activation, and moderately better modulation of systemic immune responses during long-term colitis. This may be due to the fact that BD-1 + Ceftri can deliberately prolong the colonization time of some beneficial microbiota (e.g., Bifidobacterium) and reduce the relative abundance of inflammation-related microbiota (e.g., Escherichia/Shigella and Ruminococcus). Interestingly, we found that the changes in the gut barrier and immunity were already present immediately after early intervention with BD-1 + Ceftri, implying that early effects can persist with appropriate intervention. Furthermore, intervention with BD-1 alone in early life confers an anti-inflammatory effect to a certain degree in the long-term, which may be due to the interaction between BD-1 and the host's native gut microbiota affecting intestinal metabolites. In conclusion, BD-1 was not as effective as BD-1 + Ceftri in early life, perhaps due to its failure to fully play the role of the strain itself under the influence of the host's complex microbiota. Therefore, further research is needed to explore specific mechanisms for single strain and native microbiota or the combination between probiotics and antibiotics.

8.
Nutrients ; 14(24)2022 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-36558556

RESUMO

Evidence shows that breast milk microbiota and an infant's gut microbiota are related. This study aimed to compare the effects of breast milk microbiota on the construction and colonization of gut microbiota in newborns. In this study, 23 healthy infants were selected and divided into a breastfeeding group (13) and a mixed feeding group (10) based on the feeding method within one month of age. Infant fecal and breast milk samples were collected on the day of birth (0 day) and 30 days after birth (30 days) for 16S rRNA second-generation sequencing and SCFA detection. The results showed that Firmicutes and Actinobacteriota on day 0 and Firmicutes and Proteobacteria on 30 d dominated breast milk gut microbiota. There were correlations between the breast milk microbiota and the infant gut microbiota in each group (p < 0.05). Additionally, breast milk microbiota correlated more significantly with infants' SCFAs in the breastfeeding group than in the mixed feeding group. This study showed that breast milk microbiota partially influences the construction of infant gut microbiota, with some key strains having a crucial influence, such as Lactobacillus, Bifidobacterium, and Enterobacter. However, the effect of breast milk microbiota on infant gut microbiota is not through direct strain transmission but has been indirectly influenced, which may be related to the cross-feeding effect mediated by SCFAs.


Assuntos
Microbioma Gastrointestinal , Microbiota , Feminino , Humanos , Lactente , Recém-Nascido , Leite Humano/microbiologia , Estudos de Coortes , RNA Ribossômico 16S/genética , Aleitamento Materno , Fezes/microbiologia
9.
Nutrients ; 14(17)2022 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-36079908

RESUMO

Commensal microorganisms in the human gut are a good source of candidate probiotics, particularly those with immunomodulatory effects that may improve health outcomes by regulating interactions between the gut microbiome and distal organs. Previously, we used an immune-based screening strategy to select two potential probiotic strains from infant feces in China, Bifidobacterium breve 207-1 (207-1) and Lacticaseibacillus paracasei 207-27 (207-27). In this study, the in vitro immunological effects and potential in vivo general health benefits of these two strains were evaluated using Lacticaseibacillus rhamnosus GG (LGG) as the control. The results showed that 207-1 and 207-27 significantly and differentially modulated the cytokine profiles of primary splenic cells, while did not induce abnormal systemic immune responses in healthy mice. They also modulated the gut microbiota composition in a strain-dependent manner, thus decreasing Gram-negative bacteria and increasing health-promoting taxa and short-chain fatty acid levels, particularly butyric acid. Conclusively, 207-1 and 207-27 shaped a robust gut environment in healthy mice in a strain-specific manner. Their potential immunomodulatory effects and other elite properties will be further explored using animal models of disease and subsequent clinical trials. This immune-based screening strategy is promising in efficiently and economically identifying elite candidate probiotics.


Assuntos
Microbioma Gastrointestinal , Lacticaseibacillus rhamnosus , Probióticos , Animais , Ácidos Graxos Voláteis , Fezes/microbiologia , Microbioma Gastrointestinal/fisiologia , Humanos , Lactente , Camundongos , Probióticos/farmacologia , Probióticos/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA