Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Clin Transl Sci ; 14(5): 1955-1966, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33982376

RESUMO

Metformin is a major treatment for type 2 diabetes. This study was conducted to investigate the impact of gut microbiome dysbiosis on the pharmacokinetics and antihyperglycemic effects of metformin. Healthy adult males aged 19-45 years with no defecation abnormalities were recruited for this 4-period clinical study: baseline; post-metformin (i.e., multiple oral doses of 1000 mg metformin on days 1-4); post-vancomycin (i.e., multiple oral doses of 500 mg vancomycin on days 11-17 inducing gut microbiome changes); and post-metformin + vancomycin (i.e., multiple oral doses of 1000 mg metformin on days 16-19). In each period, serum glucose and insulin concentrations following an oral glucose tolerance test, fecal samples for gut microbiome composition, and safety data were obtained. Following metformin dosing, plasma and urine samples for pharmacokinetics were collected. Nine subjects completed the study. The pharmacokinetics of metformin remained unchanged, and the antihyperglycemic effect was significantly decreased after vancomycin administration (p value = 0.039), demonstrating the weak relationship between the pharmacokinetics and pharmacodynamics of metformin. Relative abundances of some genus were changed after vancomycin administration, and tended to correlate with the antihyperglycemic effects of metformin (p value = 0.062 for Erysipelatoclostridium; p value = 0.039 for Enterobacter; and p value = 0.086 for Faecalibacterium). Adverse events occurred in all subjects and were resolved without sequelae. In conclusion, a decrease in the antihyperglycemic effect of metformin was observed after concomitant administration with vancomycin, without changes in metformin pharmacokinetics. The antihyperglycemic effect was tended to correlate with the relative abundance of several genus, suggesting that the effect of metformin is partly attributable to the gut microbiome (ClinicalTrials.gov, NCT03809260).


Assuntos
Disbiose/induzido quimicamente , Microbioma Gastrointestinal/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Vancomicina/efeitos adversos , Adulto , Diabetes Mellitus Tipo 2/tratamento farmacológico , Interações Medicamentosas , Disbiose/microbiologia , Enterobacter/efeitos dos fármacos , Enterobacter/isolamento & purificação , Faecalibacterium/efeitos dos fármacos , Faecalibacterium/isolamento & purificação , Fezes/microbiologia , Firmicutes/efeitos dos fármacos , Firmicutes/isolamento & purificação , Voluntários Saudáveis , Humanos , Hipoglicemiantes/uso terapêutico , Insulina/sangue , Masculino , Metformina/uso terapêutico , Pessoa de Meia-Idade , Vancomicina/administração & dosagem , Vancomicina/uso terapêutico , Adulto Jovem
2.
Am J Drug Alcohol Abuse ; 46(1): 4-12, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31689142

RESUMO

Background: A growing body of evidence highlights the role of the intestine in the development of various alcohol use disorder (AUD) complications. The intestinal microbiome has been proposed as an essential factor in mediating the development of AUD complications such as alcoholic liver disease.Objectives: To provide a comprehensive description of alcohol-induced intestinal microbiome alterations.Methods: We conducted a systematic review of studies investigating the effect of alcohol on the intestinal microbiome using the PRISMA checklist. We searched the Medline database on the PubMed platform for studies determining the effect of alcohol on microbiota in individuals with AUD. The manual search included references of retrieved articles. Only human studies examining the intestinal bacterial microbiome using 16S ribosomal RNA sequencing were included. Data comparing relative abundances of bacteria comprising intestinal microbiota was extracted.Results: We retrieved 17 studies investigating intestinal microbiome alterations in individuals with AUD. Intestinal microbiome alterations in individuals with AUD included depletion of Akkermansia muciniphila and Faecalibacterium prausnitzii and an increase of Enterobacteriaceae. At the phylum level, a higher abundance of Proteobacteria and lower of Bacteroidetes were found. Mixed results regarding Bifidobacterium were obtained. Several species of short-chain fatty acids producing bacteria had a lower abundance in individuals with alcohol use disorder.Conclusion: Intestinal microbiome alterations associated with dysbiosis in individuals with AUD are generally consistent across studies, making it a promising target in potential AUD complications treatment.


Assuntos
Alcoolismo/microbiologia , Fenômenos Fisiológicos Bacterianos/efeitos dos fármacos , Disbiose/microbiologia , Microbioma Gastrointestinal , Akkermansia/efeitos dos fármacos , Bactérias/classificação , Bacteroidetes/efeitos dos fármacos , Enterobacteriaceae/efeitos dos fármacos , Faecalibacterium/efeitos dos fármacos , Fibrose/microbiologia , Hepatite Alcoólica/microbiologia , Humanos , Proteobactérias/efeitos dos fármacos
3.
Food Funct ; 10(12): 7828-7843, 2019 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-31778135

RESUMO

Rhamnogalacturonan-I (RG-I)-enriched pectin (WRP) was recovered from citrus processing water by sequential acid and alkaline treatments in a previous study. RG-I-enriched pectin was proposed as a potential supplement for functional food and pharmaceutical development. However, previous studies illustrated that favorable modulations of gut microbiota by RG-I-enriched pectin were based on in vitro changes in the overall microbial structure and the question of whether there is a structure-dependent modulation of gut microbiota remains largely enigmatic. In the present study, modulations of gut microbiota by commercial pectin (CP), WRP and its depolymerized fraction (DWRP) with different RG-I contents and Mw were compared in vivo. It was revealed by 16s rRNA high-throughput sequencing that WRP and DWRP mainly composed of RG-I modulated the gut microbiota in a positive way. DWRP significantly increased the abundance of prebiotic such as Bifidobacterium spp., Lactobacillus spp., while WRP increased SCFAs producers including species in Ruminococcaceae family. By maintaining a more balanced gut microbiota composition and enriching some SCFA producers, dietary WRP and DWRP also elevated the SCFA content in the colon. Collectively, our findings offer new insights into the structure-activity correlation of citrus pectin and provide impetus towards the development of RG-I-enriched pectin with small molecular weight for specific use in health-promoting prebiotic ingredients and therapeutic products.


Assuntos
Bactérias/metabolismo , Bifidobacterium/crescimento & desenvolvimento , Ácidos Graxos Voláteis/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Lactobacillus/crescimento & desenvolvimento , Pectinas/farmacologia , Extratos Vegetais/farmacologia , Animais , Bactérias/efeitos dos fármacos , Bifidobacterium/efeitos dos fármacos , Citrus/química , Faecalibacterium/efeitos dos fármacos , Faecalibacterium/crescimento & desenvolvimento , Fermentação , Lactobacillus/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Pectinas/análise , Extratos Vegetais/análise , Prebióticos/análise
4.
Biomed Res Int ; 2019: 4530203, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31073525

RESUMO

Effects of the microbiome associated with diarrhea-predominant irritable bowel syndrome (IBS-D) on the gut have been reported, but no study has reported the effects of the IBS-D gut microbiome on the liver. We transplanted the fecal microbiota from an IBS-D patient and from a healthy volunteer to GF rats. The hepatic inflammation, serum biochemical parameters and metabolome, fecal microbiota profile, fecal short-chain fatty acids (SCFAs), and correlations among them before and after berberine intervention were assessed. Compared with the healthy control fecal microbiome transplantation (FMT) rats, the fecal microbiota of IBS-D patients induces significant Kupffer cell hyperplasia, hepatic sinusoid hypertrophy, and elevated levels of hepatic tumor necrosis factor-α and interferon-γ and decreases the synthesis of ALB in GF rats. This is possibly related to Faecalibacterium and Bifidobacterium attributable to fecal formate, acetate, and propionate levels, which are associated with the host linoleic acid pathway. Berberine can partially reverse the Kupffer cell hyperplasia, Faecalibacterium, fecal formate, acetate, and propionate by modulating the gut microbiome composition. These results may imply that IBS-D not only is an intestinal functional disorder but can cause liver inflammation, thus providing some implications regarding the clinical cognition and treatment of IBS-D.


Assuntos
Berberina/administração & dosagem , Diarreia/tratamento farmacológico , Inflamação/tratamento farmacológico , Síndrome do Intestino Irritável/tratamento farmacológico , Animais , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/patogenicidade , Diarreia/microbiologia , Diarreia/patologia , Modelos Animais de Doenças , Faecalibacterium/efeitos dos fármacos , Faecalibacterium/patogenicidade , Transplante de Microbiota Fecal , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Humanos , Inflamação/microbiologia , Inflamação/patologia , Síndrome do Intestino Irritável/microbiologia , Síndrome do Intestino Irritável/patologia , Células de Kupffer/efeitos dos fármacos , Células de Kupffer/patologia , Fígado/microbiologia , Fígado/patologia , Ratos
5.
J Microbiol ; 56(12): 886-892, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30484158

RESUMO

Statin response shows great interindividual variations. Recently, emerging studies have shown that gut microbiota is linked to therapeutic responses to drugs, including statins. However, the association between the gut bacteria composition and statin response is still unclear. In this study, gut microbiota of 202 hyperlipidemic patients with statin sensitive (SS) response and statin resistant (SR) response in East China were investigated by high throughput sequencing to compare the gut bacteria composition and biodiversity in distinct statin response patients. Higher biodiversity was detected in Group SS than Group SR. Specifically, group SS showed significantly increased proportion of genera Lactobacillus (P = 0.001), Eubacterium (P = 0.004), Faecalibacterium (P = 0.005), and Bifidobacterium (P = 0.002) and decreased proportion of genus Clostridium (P = 0.001) compared to Group SR. The results indicated that higher gut biodiversity was associated with statin sensitive response. The increased genera Lactobacillus, Eubacterium, Faecalibacterium, Bifidobacterium, and decreased genus Clostridium in patient gut microbiota may predict patient's statin response, and hence may guide statin dosage adjustments.


Assuntos
Bactérias/classificação , Bactérias/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Bactérias/genética , Bifidobacterium/efeitos dos fármacos , Biodiversidade , China , Clostridium/efeitos dos fármacos , DNA Bacteriano/genética , Eubacterium/efeitos dos fármacos , Faecalibacterium/efeitos dos fármacos , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Hipercolesterolemia/tratamento farmacológico , Lactobacillus/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade
6.
Digestion ; 97(2): 195-204, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29316555

RESUMO

BACKGROUND: Gut dysbiosis associated with the use of proton-pump inhibitors (PPIs) has been found to lead to the occurrence of infectious and inflammatory adverse events. A longitudinal observational cohort study has demonstrated the heightened risk of death associated with PPI use. SUMMARY: We evaluated meta-analyses to determine the association between PPI use and infectious and inflammatory diseases. Meta-analyses showed that PPI use is a potential risk for the development of enteric infections caused by Clostridium difficile, as well as small intestinal bacterial overgrowth, spontaneous bacterial peritonitis, community-acquired pneumonia, hepatic encephalopathy, and adverse outcomes in inflammatory bowel disease. We also examined changes in the composition and function of the gut microbiota with the use of PPIs. PPI use significantly increased the presence of Streptococcaceae and Enterococcaceae, which are risk factors for C. difficile infection, and decreased that of Faecalibacterium, a commensal anti-inflammatory microorganism. Key Message: High-throughput, microbial 16S rRNA gene sequencing has allowed us to investigate the association between the gut microbiome and PPI use. Future prospective comparison studies are necessary to confirm this association, and to develop new strategies to prevent complications of PPI use.


Assuntos
Clostridioides difficile/patogenicidade , Infecções por Clostridium/microbiologia , Disbiose/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Enteropatias/microbiologia , Inibidores da Bomba de Prótons/efeitos adversos , Clostridioides difficile/genética , Clostridioides difficile/isolamento & purificação , DNA Bacteriano/isolamento & purificação , Enterococcaceae/efeitos dos fármacos , Enterococcaceae/genética , Enterococcaceae/isolamento & purificação , Faecalibacterium/efeitos dos fármacos , Faecalibacterium/genética , Faecalibacterium/isolamento & purificação , Humanos , Intestinos/efeitos dos fármacos , Intestinos/microbiologia , Metanálise como Assunto , RNA Ribossômico 16S/genética , Fatores de Risco , Análise de Sequência de DNA , Streptococcaceae/efeitos dos fármacos , Streptococcaceae/genética , Streptococcaceae/isolamento & purificação
7.
Microbiome ; 5(1): 161, 2017 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-29262868

RESUMO

BACKGROUND: The human gut microbiome has been linked to numerous components of health and disease. However, approximately 25% of the bacterial species in the gut remain uncultured, which limits our ability to properly understand, and exploit, the human microbiome. Previously, we found that growing environmental bacteria in situ in a diffusion chamber enables growth of uncultured species, suggesting the existence of growth factors in the natural environment not found in traditional cultivation media. One source of growth factors proved to be neighboring bacteria, and by using co-culture, we isolated previously uncultured organisms from the marine environment and identified siderophores as a major class of bacterial growth factors. Here, we employ similar co-culture techniques to grow bacteria from the human gut microbiome and identify novel growth factors. RESULTS: By testing dependence of slow-growing colonies on faster-growing neighboring bacteria in a co-culture assay, eight taxonomically diverse pairs of bacteria were identified, in which an "induced" isolate formed a gradient of growth around a cultivatable "helper." This set included two novel species Faecalibacterium sp. KLE1255-belonging to the anti-inflammatory Faecalibacterium genus-and Sutterella sp. KLE1607. While multiple helper strains were identified, Escherichia coli was also capable of promoting growth of all induced isolates. Screening a knockout library of E. coli showed that a menaquinone biosynthesis pathway was required for growth induction of Faecalibacterium sp. KLE1255 and other induced isolates. Purified menaquinones induced growth of 7/8 of the isolated strains, quinone specificity profiles for individual bacteria were identified, and genome analysis suggests an incomplete menaquinone biosynthetic capability yet the presence of anaerobic terminal reductases in the induced strains, indicating an ability to respire anaerobically. CONCLUSIONS: Our data show that menaquinones are a major class of growth factors for bacteria from the human gut microbiome. These organisms are taxonomically diverse, including members of the genus Faecalibacterium, Bacteroides, Bilophila, Gordonibacter, and Sutterella. This suggests that loss of quinone biosynthesis happened independently in many lineages of the human microbiota. Quinones can be used to improve existing bacterial growth media or modulate the human gut microbiota by encouraging the growth of important symbionts, such as Faecalibacterium species.


Assuntos
Bactérias/efeitos dos fármacos , Bactérias/crescimento & desenvolvimento , Microbioma Gastrointestinal/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Vitamina K 2/metabolismo , Actinobacteria/efeitos dos fármacos , Actinobacteria/crescimento & desenvolvimento , Fenômenos Fisiológicos Bacterianos/efeitos dos fármacos , Técnicas Bacteriológicas , Técnicas de Cocultura , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Faecalibacterium/efeitos dos fármacos , Faecalibacterium/crescimento & desenvolvimento , Fezes/microbiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Filogenia , Sideróforos/metabolismo , Ubiquinona/metabolismo , Vitamina K 2/farmacologia
8.
Proc Natl Acad Sci U S A ; 114(3): E367-E375, 2017 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-28049818

RESUMO

Directed modulation of the colonic bacteria to metabolize lactose effectively is a potentially useful approach to improve lactose digestion and tolerance. A randomized, double-blind, multisite placebo-controlled trial conducted in human subjects demonstrated that administration of a highly purified (>95%) short-chain galactooligosaccharide (GOS), designated "RP-G28," significantly improved clinical outcomes for lactose digestion and tolerance. In these individuals, stool samples were collected pretreatment (day 0), after GOS treatment (day 36), and 30 d after GOS feeding stopped and consumption of dairy products was encouraged (day 66). In this study, changes in the fecal microbiome were investigated using 16S rRNA amplicon pyrosequencing and high-throughput quantitative PCR. At day 36, bifidobacterial populations were increased in 27 of 30 of GOS subjects (90%), demonstrating a bifidogenic response in vivo. Relative abundance of lactose-fermenting Bifidobacterium, Faecalibacterium, and Lactobacillus were significantly increased in response to GOS. When dairy was introduced into the diet, lactose-fermenting Roseburia species increased from day 36 to day 66. The results indicated a definitive change in the fecal microbiome of lactose-intolerant individuals, increasing the abundance of lactose-metabolizing bacteria that were responsive to dietary adaptation to GOS. This change correlated with clinical outcomes of improved lactose tolerance.


Assuntos
Microbioma Gastrointestinal/efeitos dos fármacos , Lactose/metabolismo , Oligossacarídeos/administração & dosagem , Adulto , Bifidobacterium/efeitos dos fármacos , Colo/metabolismo , Método Duplo-Cego , Faecalibacterium/efeitos dos fármacos , Fezes/microbiologia , Feminino , Humanos , Lactobacillus/efeitos dos fármacos , Masculino , RNA Ribossômico 16S/metabolismo
9.
Anaerobe ; 39: 136-42, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27018325

RESUMO

The influence of five oligosaccharides (cellobiose, stachyose, raffinose, lactulose and chito-oligosaccharides) on the adhesion of eight gut bacteria (Bifidobacterium bifidum ATCC 29521, Bacteroides thetaiotaomicron ATCC 29148D-5, Clostridium leptum ATCC 29065, Blautia coccoides ATCC 29236, Faecalibacterium prausnitzii ATCC 27766, Bacteroides fragilis ATCC 23745, Clostridium difficile ATCC 43255 and Lactobacillus casei ATCC 393) to mucous secreting and non-mucous secreting HT-29 human epithelial cells, was investigated. In pure culture, the bacteria showed variations in their ability to adhere to epithelial cells. The effect of oligosaccharides diminished adhesion and the presence of mucus played a major factor in adhesion, likely due to high adhesiveness to mucins present in the native human mucus layer covering the whole cell surface. However, clostridia displayed almost the same level of adhesion either with or without mucus being present. Bl. coccoides adhesion was decreased by stachyose and cellobiose in non-mucus-secreting cells in pure culture, while in mixed faecal culture cellobiose displayed the highest antiadhesive activity with an overall average of 65% inhibition amongst tested oligomers and lactulose displayed the lowest with an average of 47.4%. Bifidobacteria, Bacteroides, lactobacilli and clostridia were inhibited within the following ranges 47-78%, 32-65%, 11.7-58% and 64-85% respectively. This means that clostridia were the most strongly influenced members of the microflora amongst the bacterial groups tested in mixed culture. In conclusion, introducing oligosaccharides which are candidate prebiotics into pure or mixed cultures has affected bacterial adhesion.


Assuntos
Aderência Bacteriana/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Oligossacarídeos/farmacologia , Prebióticos/análise , Bacteroides/efeitos dos fármacos , Bacteroides/fisiologia , Bacteroides fragilis/efeitos dos fármacos , Bacteroides fragilis/fisiologia , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/fisiologia , Clostridium/efeitos dos fármacos , Clostridium/fisiologia , Faecalibacterium/efeitos dos fármacos , Faecalibacterium/fisiologia , Células HT29 , Humanos , Lacticaseibacillus casei/efeitos dos fármacos , Lacticaseibacillus casei/fisiologia , Mucinas/metabolismo , Especificidade da Espécie
10.
Appl Environ Microbiol ; 82(9): 2693-2699, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26921420

RESUMO

This study was performed to determine the effects of feeding a fiber-rich fraction of Brassica vegetables on the immune response through changes in enteric bacteria and short-chain fatty acid (SCFA) production in normal mice. The boiled-water-insoluble fraction of Brassica rapa L. (nozawana), which consists mainly of dietary fiber, was chosen as a test material. A total of 31 male C57BL/6J mice were divided into two groups and housed in a specific-pathogen-free facility. The animals were fed either a control diet or the control diet plus the insoluble B. rapa L. fraction for 2 weeks and sacrificed to determine microbiological and SCFA profiles in lower-gut samples and immunological molecules. rRNA-based quantification indicated that the relative population of Bacteroidetes was markedly lower in the colon samples of the insoluble B. rapa L. fraction-fed group than that in the controls. Populations of the Eubacterium rectale group and Faecalibacterium prausnitzii, both of which are representative butyrate-producing bacteria, doubled after 2 weeks of fraction intake, accompanying a marginal increase in the proportion of colonic butyrate. In addition, feeding with the fraction significantly increased levels of the anti-inflammatory cytokine interleukin-10 (IL-10) and tended to increase splenic regulatory T cell numbers but significantly reduced the population of cells expressing activation markers. We demonstrated that inclusion of the boiled-water-insoluble fraction of B. rapa L. can alter the composition of the gut microbiota to decrease the numbers of Bacteroidetes and to increase the numbers of butyrate-producing bacteria, either of which may be involved in the observed shift in the production of splenic IL-10.


Assuntos
Bactérias/metabolismo , Brassica rapa , Ácido Butírico/metabolismo , Colo/imunologia , Colo/microbiologia , Fibras na Dieta/administração & dosagem , Animais , Bactérias/efeitos dos fármacos , Bactérias/imunologia , Bactérias/isolamento & purificação , Bacteroidetes/efeitos dos fármacos , Bacteroidetes/isolamento & purificação , Bacteroidetes/metabolismo , Colo/efeitos dos fármacos , Suplementos Nutricionais , Eubacterium/efeitos dos fármacos , Eubacterium/isolamento & purificação , Eubacterium/metabolismo , Faecalibacterium/efeitos dos fármacos , Faecalibacterium/isolamento & purificação , Faecalibacterium/metabolismo , Ácidos Graxos Voláteis/biossíntese , Fezes/microbiologia , Interleucina-10/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA