Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Gut Microbes ; 11(5): 1385-1404, 2020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32515658

RESUMO

BACKGROUND: Proton pump inhibitors (PPIs) can alleviate upper gastrointestinal injury but paradoxically exacerbate aspirin (ASA)-induced small intestine injury. In this study, our goal was to simulate this exacerbation by developing an appropriate animal model, which may help in establishing treatments. Methods: Male mice were fed a 60% fructose diet for 9 weeks, then administered 200 mg/kg ASA 3 h before sacrifice. The PPI omeprazole was administered intraperitoneally once daily for 9 weeks. Bifidobacterium bifidum G9-1 was administered orally for the last week. In addition, Akkermansia muciniphila was administered orally for 9 weeks instead of omeprazole. Results: ASA-induced small-intestine injury was observed in high-fructose fed mice. Omeprazole exacerbated ASA-induced intestinal damage, significantly decreased Bifidobacteria levels, and significantly increased A. muciniphila counts in the jejunum. The direct administration of A. muciniphila caused thinning of the jejunum mucus layer, which was also observed in mice that received ASA and omeprazole. On the other hand, the administration of Bifidobacterium bifidum G9-1 inhibited A. muciniphila growth and reduced thinning of the mucus layer. The number of goblet cells in the jejunum was reduced by the administration of ASA and omeprazole, while Bifidobacterium bifidum G9-1 prevented the reduction. Conclusions: These results suggest that omeprazole-induced gut dysbiosis promotes Akkermansia growth and inhibits Bifidobacterium growth, leading to a thinning of the mucus layer through a reduction in goblet cells in the small intestine. Probiotics are, therefore, a promising approach for the treatment of small intestine injury.


Assuntos
Aspirina/efeitos adversos , Bifidobacterium bifidum , Intestino Delgado/microbiologia , Muco/metabolismo , Omeprazol/efeitos adversos , Probióticos , Akkermansia/crescimento & desenvolvimento , Akkermansia/metabolismo , Animais , Bifidobacterium bifidum/crescimento & desenvolvimento , Citocinas/metabolismo , Açúcares da Dieta/administração & dosagem , Células Caliciformes/metabolismo , Humanos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Jejuno/efeitos dos fármacos , Jejuno/microbiologia , Jejuno/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Permeabilidade , Inibidores da Bomba de Prótons/efeitos adversos , Linfócitos T Reguladores/imunologia
3.
Microbiol Immunol ; 63(12): 481-486, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31544979

RESUMO

Diarrhea is largely caused by dysbiosis accompanying the hyperproliferation of Escherichia coli (E. coli). While current treatments can resolve the symptoms, they cannot suppress the proliferation of pathogenic bacteria in the intestine. Probiotics have numerous beneficial effects on host health, including restoring the balance of the intestinal microbiota. This study investigated the effect of the probiotic Bifidobacterium bifidum G9-1 (BBG9-1), which is active in intestinal dysbiosis, in the incidence of diarrhea, in the composition of the intestinal microbiota, and in the intestinal tissue of a rat model of phytohemagglutinin (PHA)-induced diarrhea. The rats were treated with PHA, with and without BBG9-1, and the microbiota composition throughout the intestine and stool was examined using high-throughput 16S rRNA sequencing. In line with previous reports, PHA administration caused diarrhea as well as dysbiosis due to E. coli hyperproliferation. Histological findings indicated that the jejunal villus length was shortened. Rats that received BBG9-1 showed clear improvements in dysbiosis, diarrhea symptoms, and jejunal villus length. Principal coordinates analysis demonstrated the microbiota profile to be more similar between the BBG9-1 and normal groups than between the PHA and normal groups. These results indicated that BBG9-1 suppresses the hyperproliferation of E. coli and restores the jejunal villus length, thereby improving dysbiosis, and in turn, alleviating the symptoms of diarrhea.


Assuntos
Bifidobacterium bifidum/crescimento & desenvolvimento , Diarreia , Disbiose , Microbioma Gastrointestinal , Probióticos/uso terapêutico , Animais , Diarreia/tratamento farmacológico , Diarreia/microbiologia , Disbiose/tratamento farmacológico , Disbiose/microbiologia , Fezes/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Intestinos/microbiologia , Masculino , Ratos , Ratos Wistar
4.
Biosci Microbiota Food Health ; 38(2): 49-53, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31106107

RESUMO

Constipation, a functional disorder of the digestive system, is common in children and adults and may compromise patient quality of life. Because many patients are not satisfied with the efficacy of existing therapies, in this study, we investigated the efficacy of the probiotic Bifidobacterium bifidum G9-1 (BBG9-1) in constipation induced by a low-fiber diet. After inducing constipation in rats by feeding a low-fiber diet, rats were fed a low-fiber diet mixed with BBG9-1 in 14 days to determine the efficacy of BBG9-1 for alleviating constipation. BBG9-1 significantly alleviated the dysbiosis induced by a low-fiber diet and improved the fecal counts, fecal weights, and fecal water contents. Moreover, it also improved organic acid concentrations in the cecal contents. These results suggested that in low-fiber diet-induced constipation, BBG9-1 could alleviate dysbiosis and constipation and may improve the intestinal environment, supporting its potential application in the treatment of constipation.

5.
Sci Rep ; 8(1): 12384, 2018 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-30120330

RESUMO

Although gut microbiota and early life events are likely involved in the development of irritable bowel syndrome (IBS), it remains unclear how these factors interact in the pathophysiology of IBS. In the present study, using rats subjected to maternal separation (MS) as a model of IBS, we investigated interrelationships among gut microbiota, stress susceptibility and intestinal permeability, and examined the effect of the probiotic Bifidobacterium bifidum G9-1 (BBG9-1) on those interrelationships. When compared with the controls at postnatal day 20, MS rats showed hypercorticosteronemia, enhanced intestinal permeability and changes in gut microbiota structure. All of these changes in MS rats were prevented by treatment with BBG9-1. Although the gut microbiota profile and basal serum corticosterone level did not differ between MS and control rats at postnatal day 56, MS rats showed hypersensitivity to restraint stress in terms of serum corticosterone level and fecal frequency. However, such hypersensitivity was not observed in MS rats treated with BBG9-1. These findings suggest that MS initiates the link between gut microbiota alteration and hypersensitivity to stress and that the triggering of this process can be prevented by the treatment with the probiotic BBG9-1.


Assuntos
Bifidobacterium bifidum , Biodiversidade , Microbioma Gastrointestinal , Probióticos , Estresse Fisiológico , Animais , Biomarcadores , Peso Corporal , Corticosterona/sangue , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Permeabilidade , Ratos
6.
Basic Clin Pharmacol Toxicol ; 121(3): 159-168, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28374966

RESUMO

The chemotherapeutic agent 5-fluorouracil (5-FU) causes intestinal mucositis with severe diarrhoea, but the pathogenesis is not fully understood. In this study, we investigated the pathogenic effects of 5-FU in mice, focusing on apoptosis, enterobacteria and inflammatory cytokines. Repeated administration of 5-FU caused severe intestinal mucositis on day 6, accompanied by diarrhoea and body-weight loss. TNF-α expression increased 1 day after exposure to the drug, and spiked a second time on day 4, at which point myeloperoxidase activity and IL-1ß expression also increased. Apoptotic cells were observed in intestinal crypts only on day 1. 5-FU also induced dysbiosis, notably decreasing the abundance of intestinal Firmicutes while increasing the abundance of Bacteroidetes and Verrucomicrobia. Twice-daily co-administration of oral antibiotics significantly reduced the severity of intestinal mucositis and dysbiosis, and blocked the increase in myeloperoxidase activity and cytokine expression on day 6, without affecting apoptosis and TNF-α up-regulation on day 1. In cultured colonic epithelial cells, exposure to 5-FU also up-regulated TNF-α expression. Collectively, the data suggest that crypt apoptosis, dysbiosis and expression of inflammatory cytokines are sequential events in the development of intestinal mucositis after exposure to 5-FU. In particular, 5-FU appears to directly induce apoptosis via TNF-α and to suppress intestinal cell proliferation, thereby resulting in degradation of the epithelial barrier, as well as in secondary inflammation mediated by inflammatory cytokines.


Assuntos
Antimetabólitos Antineoplásicos/efeitos adversos , Apoptose/efeitos dos fármacos , Citocinas/metabolismo , Disbiose/etiologia , Fluoruracila/efeitos adversos , Enteropatias/induzido quimicamente , Mucosite/induzido quimicamente , Animais , Antibacterianos/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Citocinas/genética , Diarreia/etiologia , Diarreia/imunologia , Diarreia/microbiologia , Diarreia/prevenção & controle , Quimioterapia Combinada , Disbiose/imunologia , Disbiose/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Negativas/crescimento & desenvolvimento , Bactérias Gram-Negativas/imunologia , Enteropatias/metabolismo , Enteropatias/patologia , Enteropatias/fisiopatologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Cinética , Masculino , Camundongos Endogâmicos C57BL , Mucosite/metabolismo , Mucosite/patologia , Mucosite/fisiopatologia , Redução de Peso
7.
PLoS One ; 12(3): e0173979, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28346473

RESUMO

Human rotavirus (RV) infection is a leading cause of dehydrating diarrhea in infants and young children worldwide. Since therapeutic approaches to RV gastroenteritis are limited to alleviation of dehydration with oral rehydration solutions, more direct approaches to palliate symptoms of RV gastroenteritis are required. Treatments with probiotics have been increasingly recognized as alternative safe and low cost treatments for moderate infectious diarrhea. In this study, Bifidobacterium bifidum G9-1 (BBG9-1), which has been used as an intestinal drug for several decades, was shown to have a remarkable protective effect against RV gastroenteritis in a suckling mice model. As well as prophylactic oral administration of BBG9-1 from 2 days before RV infection, therapeutic oral administration of BBG9-1 from 1 day after RV infection significantly alleviated RV-induced diarrhea. Therapeutic administration of BBG9-1 reduced various types of damage in the small intestine, such as epithelial vacuolization and villous shortening, and significantly diminished the infectious RV titer in mixtures of cecal contents and feces. It was also shown that therapeutic administration of BBG9-1 significantly increased the number of acidic mucin-positive goblet cells and the gene expression of mucosal protective factors including MUC2, MUC3, MUC4, TGFß1 and TFF3 in the small intestine. This led to alleviation of low gut permeability shown as decreased gene expression levels of occludin, claudin-1 and villin-1 after RV infection. Furthermore, in the small intestine, therapeutic administration of BBG9-1 significantly palliated the decreased gene expression of SGLT-1, which plays an important role in water absorption. In the large intestine, administered BBG9-1 was shown to replicate to assimilate undigested nutrients, resulting in normalization of the abnormally high osmotic pressure. These results suggested that water malabsorption caused by RV infection was alleviated in mice administered BBG9-1. Thus, the present study showed that oral administration of BBG9-1 palliated diarrhea partly through protection against RV-induced lesions by inducing mucosal protective factors. Oral administration of BBG9-1 is thought to be an efficient method for management of an RV epidemic for both prophylactic and therapeutic purposes.


Assuntos
Bifidobacterium bifidum/fisiologia , Gastroenterite/terapia , Intestinos/microbiologia , Intestinos/patologia , Probióticos/uso terapêutico , Infecções por Rotavirus/terapia , Administração Oral , Animais , Linhagem Celular , Chlorocebus aethiops , Diarreia/complicações , Diarreia/microbiologia , Diarreia/patologia , Diarreia/terapia , Gastroenterite/complicações , Gastroenterite/microbiologia , Gastroenterite/patologia , Regulação da Expressão Gênica , Macaca mulatta , Camundongos Endogâmicos BALB C , Probióticos/administração & dosagem , Rotavirus/isolamento & purificação , Rotavirus/fisiologia , Infecções por Rotavirus/complicações , Infecções por Rotavirus/microbiologia , Infecções por Rotavirus/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA