Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 634
Filtrar
1.
BMC Genomics ; 25(1): 718, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39054474

RESUMEN

INTRODUCTION: Our understanding of particular gut microbiota members such as Bifidobacterium and Enterococcus in low-middle-income countries remains very limited, particularly early life strain-level beneficial traits. This study addresses this gap by exploring a collection of bacterial strains isolated from the gut of Zimbabwean infants; comparing their genomic characteristics with strains isolated from infants across North America, Europe, and other regions of Africa. MATERIALS AND METHOD: From 110 infant stool samples collected in Harare, Zimbabwe, 20 randomly selected samples were used to isolate dominant early-life gut microbiota members Bifidobacterium and Enterococcus. Isolated strains were subjected to whole genome sequencing and bioinformatics analysis including functional annotation of carbohydrates, human milk oligosaccharide (HMO) and protein degradation genes and clusters, and the presence of antibiotic resistance genes (ARGs). RESULTS: The study observed some location-based clustering within the main five identified taxonomic groups. Furthermore, there were varying and overall species-specific numbers of genes belonging to different GH families encoded within the analysed dataset. Additionally, distinct strain- and species-specific variances were identified in the potential of Bifidobacterium for metabolizing HMOs. Analysis of putative protease activity indicated a consistent presence of gamma-glutamyl hydrolases in Bifidobacterium, while Enterococcus genomes exhibited a high abundance of aspartyl peptidases. Both genera harboured resistance genes against multiple classes of antimicrobial drugs, with Enterococcus genomes containing a higher number of ARGs compared to Bifidobacterium, on average. CONCLUSION: This study identified promising probiotic strains within Zimbabwean isolates, offering the potential for early-life diet and microbial therapies. However, the presence of antibiotic resistance genes in infant-associated microbes raises concerns for infection risk and next-stage probiotic development. Further investigation in larger cohorts, particularly in regions with limited existing data on antibiotic and probiotic use, is crucial to validate these initial insights. IMPACT STATEMENT: This research represents the first investigation of its kind in the Zimbabwean context, focusing on potential probiotic strains within the early-life gut microbiota. By identifying local probiotic strains, this research can contribute to the development of probiotic interventions that are tailored to the Zimbabwean population, which can help address local health challenges and promote better health outcomes for infants. Another essential aspect of the study is the investigation of antimicrobial resistance genes present in Zimbabwean bacterial strains. Antimicrobial resistance is a significant global health concern, and understanding the prevalence and distribution of resistance genes in different regions can help inform public health policies and interventions.


Asunto(s)
Bifidobacterium , Enterococcus , Microbioma Gastrointestinal , Humanos , Zimbabwe , Lactante , Microbioma Gastrointestinal/genética , Enterococcus/genética , Enterococcus/efectos de los fármacos , Enterococcus/aislamiento & purificación , Bifidobacterium/genética , Bifidobacterium/aislamiento & purificación , Bifidobacterium/efectos de los fármacos , Genómica , Genoma Bacteriano , Heces/microbiología , Secuenciación Completa del Genoma , Estudios de Cohortes , Filogenia
2.
Nutrients ; 16(13)2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38999750

RESUMEN

(1) Background: Recently, academic studies are demonstrating that the cholesterol-lowering effects of pectin oligosaccharides (POSs) are correlated to intestinal flora. However, the mechanisms of POS on cholesterol metabolisms are limited, and the observations of intestinal flora are lacking integrative analyses. (2) Aim and methods: To reveal the regulatory mechanisms of POS on cholesterol metabolism via an integrative analysis of the gut microbiota, the changes in gut microbiota structure and metabolite composition after POS addition were investigated using Illumina MiSeq sequencing and non-targeted metabolomics through in vitro gut microbiota fermentation. (3) Results: The composition of fecal gut flora was adjusted positively by POS. POS increased the abundances of the cholesterol-related bacterial groups Bacteroidetes, Bifidobacterium and Lactobacillus, while it decreased conditional pathogenic Escherichia coli and Enterococcus, showing good prebiotic activities. POS changed the composition of gut microbiota fermentation metabolites (P24), causing significant changes in 221 species of fermentation metabolites in a non-targeted metabolomics analysis and promoting the production of short-chain fatty acids. The abundances of four types of cholesterol metabolism-related metabolites (adenosine monophosphate, cyclic adenosine monophosphate, guanosine and butyrate) were significantly higher in the P24 group than those in the control group without POS addition. (4) Conclusion: The abovementioned results may explain the hypocholesterolemic effects of POS and promotion effects on cholesterol efflux of P24. These findings indicated that the potential regulatory mechanisms of citrus POS on cholesterol metabolism are modulated by cholesterol-related gut microbiota and specific metabolites.


Asunto(s)
Colesterol , Heces , Fermentación , Microbioma Gastrointestinal , Oligosacáridos , Pectinas , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Pectinas/farmacología , Pectinas/metabolismo , Colesterol/metabolismo , Oligosacáridos/farmacología , Heces/microbiología , Humanos , Prebióticos , Masculino , Metabolómica , Ácidos Grasos Volátiles/metabolismo , Bifidobacterium/metabolismo , Bifidobacterium/efectos de los fármacos , Femenino , Bacterias/metabolismo , Bacterias/efectos de los fármacos , Bacterias/clasificación , Citrus
3.
Nutrients ; 16(11)2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38892631

RESUMEN

This study investigated the effect of astragalus polysaccharide (APS, an ingredient with hypoglycemic function in a traditional Chinese herbal medicine) on gut microbiota and metabolites of type 2 diabetes mellitus (T2DM) patients using a simulated fermentation model in vitro. The main components of APS were isolated, purified, and structure characterized. APS fermentation was found to increase the abundance of Lactobacillus and Bifidobacterium and decrease the Escherichia-Shigella level in the fecal microbiota of T2DM patients. Apart from increasing propionic acid, APS also caused an increase in all-trans-retinoic acid and thiamine (both have antioxidant properties), with their enrichment in the KEGG pathway associated with thiamine metabolism, etc. Notably, APS could also enhance fecal antioxidant properties. Correlation analysis confirmed a significant positive correlation of Lactobacillus with thiamine and DPPH-clearance rate, suggesting the antioxidant activity of APS was related to its ability to enrich some specific bacteria and upregulate their metabolites.


Asunto(s)
Antioxidantes , Planta del Astrágalo , Diabetes Mellitus Tipo 2 , Heces , Fermentación , Microbioma Gastrointestinal , Polisacáridos , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Polisacáridos/farmacología , Planta del Astrágalo/química , Heces/microbiología , Antioxidantes/farmacología , Masculino , Femenino , Persona de Mediana Edad , Tiamina/farmacología , Tiamina/metabolismo , Bifidobacterium/metabolismo , Bifidobacterium/efectos de los fármacos , Lactobacillus/metabolismo , Lactobacillus/efectos de los fármacos , Hipoglucemiantes/farmacología
4.
Int J Biol Macromol ; 273(Pt 1): 133053, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38857723

RESUMEN

Trehalose (α-d-glucopyranosyl-(1-1)-α-D-glucopyranoside) has found applications in diverse food products as a sweetener, stabilizer, and humectant. Recent attention has focused on trehalose due to its contradictory effects on the virulence of Clostridium difficile. In this study, we investigate the impact of novel trehalose-derived galactooligosaccharides (Treh-GOS) on the human gut microbiota using in vitro fecal fermentation models. Distinct Treh-GOS structures elicit varying taxonomic responses. For instance, ß-Gal-(1-4)-trehalose [DP3(1-4)] leads to an increase of Bifidobacterium, comparable to results observed with commercial GOS. Conversely, ß-Gal-(1-6)-trehalose [DP3(1-6)] prompts an increase in Lactobacillus. Notably, both of these trisaccharides yield the highest concentrations of butyric acid across all samples. On the other hand, Treh-GOS tetrasaccharide mixture (DP4), featuring a novel trehalose galactosylation in both glucose units, fosters the growth of Parabacteroides. Our findings underscore the capacity of novel Treh-GOS to modulate the human gut microbiota. Consequently, these innovative galactooligosaccharides emerge as promising candidates for novel prebiotic applications.


Asunto(s)
Fermentación , Microbioma Gastrointestinal , Oligosacáridos , Trehalosa , Trehalosa/farmacología , Trehalosa/química , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Oligosacáridos/farmacología , Oligosacáridos/química , Fermentación/efectos de los fármacos , Heces/microbiología , Prebióticos , Bifidobacterium/efectos de los fármacos , Bifidobacterium/metabolismo
5.
Gut Microbes ; 16(1): 2357176, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38798019

RESUMEN

Resistance to antibiotics in newborns is a huge concern as their immune system is still developing, and infections and resistance acquisition in early life have short- and long-term consequences for their health. Bifidobacterium species are important commensals capable of dominating the infant gut microbiome and are known to be less prone to possess antimicrobial resistance genes than other taxa that may colonize infants. We aimed to study the association between Bifidobacterium-dominated infant gut microbiota and the antibiotic resistant gene load in neonates, and to ascertain the perinatal factors that may contribute to the antibiotic resistance acquisition. Two hundred infant fecal samples at 7 days and 1 month of age from the MAMI birth cohort were included in the study and for whom maternal-neonatal clinical records were available. Microbiota profiling was carried out by 16S rRNA amplicon sequencing, and targeted antibiotic resistance genes (ARGs) including tetM, tetW, tetO, blaTEM, blaSHV and ermB were quantified by qPCR. Infant microbiota clustered into two distinct groups according to their Bifidobacterium genus abundance: high and low. The main separation of groups or clusters at each time point was performed with an unsupervised non-linear algorithm of k-means partitioning to cluster data by time points based on Bifidobacterium genus relative abundance. Microbiota composition differed significantly between both groups, and specific bifidobacterial species were enriched in each cluster. Lower abundance of Bifidobacterium in the infant gut was associated with a higher load of antibiotic resistance genes. Our results highlight the relevance of Bifidobacterium genus in the early acquisition and establishment of antibiotic resistance in the gut. Further studies are needed to develop strategies to promote a healthy early colonization and fight against the spread of antibiotic resistances.


Asunto(s)
Antibacterianos , Bifidobacterium , Farmacorresistencia Bacteriana , Heces , Microbioma Gastrointestinal , ARN Ribosómico 16S , Humanos , Bifidobacterium/genética , Bifidobacterium/efectos de los fármacos , Bifidobacterium/aislamiento & purificación , Recién Nacido , Microbioma Gastrointestinal/efectos de los fármacos , Heces/microbiología , Antibacterianos/farmacología , Femenino , ARN Ribosómico 16S/genética , Farmacorresistencia Bacteriana/genética , Masculino , Lactante
6.
Biomed Pharmacother ; 174: 116561, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38593705

RESUMEN

Pectin and its derivatives have been shown to modulate immune signaling as well as gut microbiota in preclinical studies, which may constitute the mechanisms by which supplementation of specific pectic polysaccharides confers protection against viral respiratory infections. In a double-blind, placebo-controlled rhinovirus (RV16) challenge study, healthy volunteers were randomized to consume placebo (0.0 g/day) (N = 46), low-dose (0.3 g/day) (N = 49) or high-dose (1.5 g/day) (N = 51) of carrot derived rhamnogalacturonan-I (cRG-I) for eight weeks and they were subsequently challenged with RV-16. Here, the effect of 8-week cRG-I supplementation on the gut microbiota was studied. While the overall gut microbiota composition in the population was generally unaltered by this very low dose of fibre, the relative abundance of Bifidobacterium spp. (mainly B. adolescentis and B. longum) was significantly increased by both doses of cRG-1. Moreover, daily supplementation of cRG-I led to a dose-dependent reduction in inter- and intra-individual microbiota heterogeneity, suggesting a stabilizing effect on the gut microbiota. The severity of respiratory symptoms did not directly correlate with the cRG-I-induced microbial changes, but several dominant groups of the Ruminococcaceae family and microbiota richness were positively associated with a reduced and hence desired post-infection response. Thus, the present results on the modulation of the gut microbiota composition support the previously demonstrated immunomodulatory and protective effect of cRG-I during a common cold infection.


Asunto(s)
Suplementos Dietéticos , Microbioma Gastrointestinal , Voluntarios Sanos , Pectinas , Humanos , Pectinas/administración & dosificación , Pectinas/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Masculino , Adulto , Método Doble Ciego , Femenino , Adulto Joven , Rhinovirus/efectos de los fármacos , Persona de Mediana Edad , Heces/microbiología , Bifidobacterium/efectos de los fármacos
7.
Benef Microbes ; 14(4): 317-334, 2023 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-38661345

RESUMEN

Recently there is much debate in the scientific community over the impact of the food matrix on prebiotic efficacy of inulin-type fructans. Previous studies suggest that prebiotic selectivity of inulin-type fructans towards bifidobacteria is unaffected by the food matrix. Due to differences in study design, definitive conclusions cannot be drawn from these findings with any degree of certainty. In this randomised trial, we aimed to determine the effects that different food matrices had on the prebiotic efficacy of inulin-type fructans following a standardised 10-day, 4-arm, parallel, randomised protocol with inulin either in pure form or incorporated into shortbread biscuits, milk chocolate or a rice drink. Similar increases in Bifidobacterium counts were documented across all four interventions using both fluorescence in situ hybridisation (pure inulin: +0.63; shortbread: +0.59; milk chocolate: +0.65 and rice drink: +0.71 (log10 cells/g wet faeces) and 16S rRNA sequencing quantitative microbiome profiling data (pure inulin: +1.21 × 109; shortbread: +1.47 × 109; milk chocolate: +8.59 × 108 and rice drink: +1.04 × 109 (cells/g wet faeces) (all P ≤ 0.05). From these results, we can confirm that irrespective of the food matrix, the selectivity of inulin-type fructans towards Bifidobacterium is unaffected, yet the compositional make-up of the food matrix may have implications regarding wider changes in the microbiota.


Asunto(s)
Bifidobacterium , Heces , Fructanos , Inulina , Prebióticos , ARN Ribosómico 16S , Inulina/farmacología , Humanos , Bifidobacterium/genética , Bifidobacterium/efectos de los fármacos , Heces/microbiología , Fructanos/farmacología , ARN Ribosómico 16S/genética , Oryza , Femenino , Masculino , Microbioma Gastrointestinal/efectos de los fármacos , Adulto , Chocolate , Adulto Joven , Hibridación Fluorescente in Situ
8.
Nutrients ; 14(2)2022 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-35057489

RESUMEN

Constipation is a major issue for 10-20% of the global population. In a double-blind randomized placebo-controlled clinical trial, we aimed to determine a dose-response effect of galacto-oligosaccharides (GOS) on stool characteristics and fecal microbiota in 132 adults with self-reported constipation according to Rome IV criteria (including less than three bowel movements per week). Subjects (94% females, aged: 18-59 years) received either 11 g or 5.5 g of BiotisTM GOS, or a control product, once daily for three weeks. Validated questionnaires were conducted weekly to study primarily stool frequency and secondary stool consistency. At base- and endline, stool samples were taken to study fecal microbiota. A trend towards an increased stool frequency was observed after the intervention with 11 g of GOS compared to control. While during screening everybody was considered constipated, not all subjects (n = 78) had less than three bowel movements per week at baseline. In total, 11 g of GOS increased stool frequency compared to control in subjects with a low stool frequency at baseline (≤3 bowel movements per week) and in self-reported constipated adults 35 years of age or older. A clear dose-response of GOS was seen on fecal Bifidobacterium, and 11 g of GOS significantly increased Anaerostipes hadrus. In conclusion, GOS seems to be a solution to benefit adults with a low stool frequency and middle-aged adults with self-reported constipation.


Asunto(s)
Estreñimiento/microbiología , Defecación/efectos de los fármacos , Heces/microbiología , Galactosa/farmacología , Oligosacáridos/farmacología , Prebióticos/administración & dosificación , Adolescente , Adulto , Bifidobacterium/efectos de los fármacos , Estreñimiento/terapia , Método Doble Ciego , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Autoinforme , Adulto Joven
9.
Front Immunol ; 12: 772532, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34970262

RESUMEN

Emerging evidence supports that the efficacy of immune checkpoint blockade (ICB) therapy is associated with the host's gut microbiota, as prior antibiotic intake often leads to poor outcome and low responsiveness toward ICB treatment. Therefore, we hypothesized that the efficacy of ICB therapy like anti-programmed cell death protein-1 (PD-1) treatment required an intact host gut microbiota, and it was established that probiotics could enhance the recovery of gut microbiota disruption by external stimuli. Thus, the present study aimed to evaluate the effect of the probiotics, Lactobacillus rhamnosus Probio-M9, on recovering antibiotic-disrupted gut microbiota and its impact on the outcome of ICB therapy in tumor-bearing mice. We first disrupted the mouse microbiota by antibiotics and then remediated the gut microbiota by probiotics or naturally. Tumor transplantation was then performed, followed by anti-PD-1-based antitumor therapy. Changes in the fecal metagenomes and the tumor suppression effect were monitored during different stages of the experiment. Our results showed that Probio-M9 synergized with ICB therapy, significantly improving tumor inhibition compared with groups not receiving the probiotic treatment (P < 0.05 at most time points). The synergistic effect was accompanied by effective restoration of antibiotic-disrupted fecal microbiome that was characterized by a drastically reduced Shannon diversity value and shifted composition of dominating taxa. Moreover, probiotic administration significantly increased the relative abundance of beneficial bacteria (e.g., Bifidobacterium pseudolongum, Parabacteroides distasonis, and some Bacteroides species; 0.0001 < P < 0.05). The gut microbiome changes were accompanied by mild reshaping of the functional metagenomes characterized by enrichment in sugar degradation and vitamin and amino acid synthesis pathways. Collectively, this study supported that probiotic administration could enhance the efficacy and responsiveness of anti-PD-1-based immunotherapy, and Probio-M9 could be a potential candidate of microbe-based synergistic tumor therapeutics. The preclinical data obtained here would support the design of future human clinical trials for further consolidating the current findings and for safety assessment of probiotic adjunctive treatment in ICB therapy.


Asunto(s)
Antibacterianos/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Lacticaseibacillus rhamnosus , Neoplasias/terapia , Probióticos/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Bacteroides/efectos de los fármacos , Bacteroides/crecimiento & desarrollo , Bifidobacterium/efectos de los fármacos , Bifidobacterium/crecimiento & desarrollo , Línea Celular Tumoral , Heces/microbiología , Ratones Endogámicos BALB C , Neoplasias/microbiología
10.
Nutrients ; 13(11)2021 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-34836092

RESUMEN

BACKGROUND: Human milk oligosaccharide supplementation safely modulates fecal bifidobacteria abundance and holds the potential to manage symptoms in irritable bowel syndrome (IBS). Here, we aimed to determine the role of a 4:1 mix of 2'-O-fucosyllactose and lacto-N-neotetraose (2'FL/LNnT) on the modulation of the gut microbiota composition and host mucosal response, as well as the link between the bifidobacteria abundance and metabolite modulation, in IBS patients. METHODS: Biological samples were collected from IBS patients (n = 58) at baseline and week 4 post-supplementation with placebo, 5 g or 10 g doses of 2'FL/LNnT. The gut microbiota composition, metabolite profiles and expression of genes related to host mucosal response were determined. RESULTS: Moderate changes in fecal, but not mucosal, microbial composition (ß-diversity) was observed during the intervention with higher dissimilarity observed within individuals receiving 10g 2'FL/LNnT compared to placebo. Both fecal and mucosal Bifidobacterium spp. increased after 2'FL/LNnT intake, with increased proportions of Bifidobacterium adolescentis and Bifidobacterium longum. Moreover, the intervention modulated the fecal and plasma metabolite profiles, but not the urine metabolite profile or the host mucosal response. Changes in the metabolite profiles were associated to changes in bifidobacteria abundance. CONCLUSION: Supplementation with 2'FL/LNnT modulated the gut microbiota, fecal and plasma metabolite profiles, but not the host mucosal response in IBS. Furthermore, the bifidogenic effect was associated with metabolite modulation. Overall, these findings support the assertion that 2'FL/LNnT supplementation modulate the intestinal microenvironment of patients with IBS, potentially related to health.


Asunto(s)
Suplementos Dietéticos , Microbioma Gastrointestinal/efectos de los fármacos , Síndrome del Colon Irritable/tratamiento farmacológico , Leche Humana/química , Oligosacáridos/farmacología , Adolescente , Adulto , Anciano , Bifidobacterium/efectos de los fármacos , Método Doble Ciego , Heces/microbiología , Femenino , Humanos , Mucosa Intestinal/efectos de los fármacos , Síndrome del Colon Irritable/microbiología , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Trisacáridos/farmacología , Adulto Joven
11.
Nutrients ; 13(11)2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34836175

RESUMEN

A high-salt diet (HSD) is one of the key risk factors for hypertension and kidney injury. In this study, a HSD C57BL/6J mice model was established with 4% NaCl, and then different concentrations of Lactobacillus plantarum ZDY2013 were intragastrically administered for 2 weeks to alleviate HSD-induced renal injury. For the study, 16S rRNA gene sequencing, non-targeted metabonomics, real-time fluorescent quantitative PCR, and Masson's staining were used to investigate the mechanism of L. plantarum ZDY2013 in alleviating renal damage. Results showed that HSD caused intestinal inflammation and changed the intestinal permeability of mice, disrupted the balance of intestinal flora, and increased toxic metabolites (tetrahydrocorticosteron (THB), 3-methyhistidine (3-MH), creatinine, urea, and L-kynurenine), resulting in serious kidney damage. Interestingly, L. plantarum ZDY2013 contributed to reconstructing the intestinal flora of mice by increasing the level of Lactobacillus and Bifidobacterium and decreasing that of Prevotella and Bacteroides. Moreover, the reconstructed intestinal microbiota significantly changed the concentration of the metabolites of hosts through metabolic pathways, including TCA cycle, ABC transport, purine metabolism, and histidine metabolism. The content of uremic toxins such as L-kynurenine, creatinine, and urea in the serum of mice was found to be decreased by L. plantarum ZDY2013, which resulted in renal injury alleviation. Our data suggest that L. plantarum ZDY2013 can indeed improve chronic kidney injury by regulating intestinal flora, strengthening the intestinal barrier, limiting inflammatory response, and reducing uremic toxins.


Asunto(s)
Enfermedades Renales/tratamiento farmacológico , Riñón/lesiones , Lactobacillus plantarum , Probióticos/farmacología , Cloruro de Sodio Dietético/efectos adversos , Animales , Bifidobacterium/efectos de los fármacos , Dieta/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Inflamación/etiología , Inflamación/metabolismo , Intestinos/metabolismo , Enfermedades Renales/etiología , Enfermedades Renales/metabolismo , Lactobacillus/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Prevotella/efectos de los fármacos , ARN Ribosómico 16S/metabolismo
12.
Nutrients ; 13(11)2021 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-34836220

RESUMEN

Grapes provide a rich source of polyphenols and fibers. This study aimed to evaluate the effect of the daily consumption of 46 g of whole grape powder, providing the equivalent of two servings of California table grapes, on the gut microbiome and cholesterol/bile acid metabolism in healthy adults. This study included a 4-week standardization to a low-polyphenol diet, followed by 4 weeks of 46 g of grape powder consumption while continuing the low-polyphenol diet. Compared to the baseline, 4 weeks of grape powder consumption significantly increased the alpha diversity index of the gut microbiome. There was a trend of increasing Verrucomicrobia (p = 0.052) at the phylum level, and a significant increase in Akkermansia was noted. In addition, there was an increase in Flavonifractor and Lachnospiraceae_UCG-010, but a decrease in Bifidobacterium and Dialister at the genus level. Grape powder consumption significantly decreased the total cholesterol by 6.1% and HDL cholesterol by 7.6%. There was also a trend of decreasing LDL cholesterol by 5.9%, and decreasing total bile acid by 40.9%. Blood triglyceride levels and body composition were not changed by grape powder consumption. In conclusion, grape powder consumption significantly modified the gut microbiome and cholesterol/bile acid metabolism.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colesterol/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Extractos Vegetales/administración & dosificación , Vitis/química , Adulto , Akkermansia/efectos de los fármacos , Bifidobacterium/efectos de los fármacos , Colesterol/sangre , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Polifenoles/metabolismo , Polvos , Triglicéridos/sangre , Verrucomicrobia/efectos de los fármacos , Adulto Joven
13.
FASEB J ; 35(11): e21977, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34613640

RESUMEN

Xylo-oligosaccharide (XOS), which is considered as a potential prebiotic, exhibits multiple beneficial effects on modulation of gut microbiota, strength of intestinal barrier, and inhibition of intestinal inflammation. The objective of this study is to investigate whether XOS protects against Salmonella infection by modulating gut microbiota, enhancing the intestinal barrier, and resisting colonization. C57BL/6 male mice received water supplementation with 5% XOS for 14 days before Salmonella Typhimurium infection. The results showed that XOS suppressed the Salmonella-induced inflammation, but had limited effects on tight junction molecules and mRNA expression of mucus proteins, except for claudin-1 in the colon. Data of 16S rDNA sequencing indicated that XOS modulated gut microbiota composition by significantly stimulating Bifidobacterium animalis (B. animalis), and reducing Salmonella counts. Therefore, the potential protective effects of B. animalis against Salmonella challenge were investigated as well. Bifidobacterium animalis subsp lactis BB-12 (BB12), which could markedly increase in XOS, was selected to treat mice. Similarly, Salmonella-induced inflammatory reactions were alleviated by BB12 but tight junction molecules and mucin proteins in the colonic tissues were not affected. Administration of BB12 remarkably decreased the copies of Salmonella in cecal digesta post Salmonella infection. Additionally, the decrease concentrations of cecal propionate and total short-chain fatty acids (SCFAs) in Salmonella-infected mice were reversed by BB12 treatment, and propionate performed a strong inhibitory effect on Salmonella growth in vitro. Besides that, BB12 could directly restrict Salmonella proliferation in vitro. Moreover, BB12 reduced the adhesion ability of Salmonella on the Caco-2 cells model. Our results suggest that XOS could be considered as a candidate of functional food to protect against Salmonella infection by stimulating Bifidobacterium, which then resists Salmonella colonization by maintaining the intestinal SCFAs levels and suppressing adhesibility.


Asunto(s)
Bifidobacterium/efectos de los fármacos , Inflamación/tratamiento farmacológico , Probióticos , Infecciones por Salmonella/tratamiento farmacológico , Salmonella typhimurium/efectos de los fármacos , Xilosa , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Intestinos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Probióticos/farmacología , Probióticos/uso terapéutico , Xilosa/análogos & derivados , Xilosa/farmacología , Xilosa/uso terapéutico
14.
Carbohydr Polym ; 273: 118546, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34560958

RESUMEN

This study investigated the effect of konjac glucomannan (KGM) of different molecular weight on fecal microflora against antibiotic disturbance. KGM (~1.8 × 107 Da) was partially hydrolysed with trifluoroacetic acid (TFA) for 10 and 60 min to KGM1 (~2.1 × 104 Da) and KGM2 (7413 Da), respectively. The acid treatment caused significant reduction of intrinsic viscosity, average molecular weight (MW) and particle size of KGM, but brought limited change to the molecular structure. Low-MW KGM2 showed the most significant effect on fecal microflora in the presence of two common antibiotics (ampicillin and clindamycin), by increasing the relative abundance of Bifidobacteriaceae while decreasing the proportion of Enterobacteriaceae. Additionally, both the native and acid-treated KGM counteracted the adverse influence of antibiotics on the production of short chain fatty acids. The results have demonstrated the effect of KGM on gut microbiota with antibiotic disturbance.


Asunto(s)
Antibacterianos/farmacología , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Mananos/farmacología , Amorphophallus/química , Antibacterianos/efectos adversos , Bifidobacterium/efectos de los fármacos , Enterobacteriaceae/efectos de los fármacos , Ácidos Grasos Volátiles/metabolismo , Fermentación , Humanos , Hidrólisis , Mananos/química , Estructura Molecular , Peso Molecular , Tamaño de la Partícula , Ácido Trifluoroacético/química , Viscosidad
15.
Int J Biol Macromol ; 188: 586-594, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34403670

RESUMEN

The exopolysaccharide (EPS) from the mycelial fermentation of a medicinal fungus Cordyceps sinensis Cs-HK1 had shown significant anti-inflammatory activity previously, and EPS-LM was a highly active fraction with a relatively low molecular weight (MW) isolated from the Cs-HK1 EPS. This study was to assess the effects of Bifidobacterial fermentation in anaerobic conditions on the molecular properties and anti-inflammatory activity of EPS-LM. In both Bifidobacterial cultures (B. breve and B. longum), EPS-LM was fractionally consumed as a carbon source, increasing the bacterial growth and acetic acid production. Analytical results from the fermentation digesta (supernatant) suggested that EPS-LM was partially degraded to lower molecular weight (MW) products with modified structures during the Bifidobacterial fermentation. More interestingly, the higher MW digesta fraction containing the partially degraded EPS-LM showed even stronger inhibiting activity than the original EPS-LM on the LPS-induced pro-inflammatory responses in THP-1 cell culture, including NF-κB activation, release of NO, TNF-α and IL-8. The study has shown that the fermentation by selected Bifidobacterial strains is effective to modify natural polysaccharides with enhanced bioactivities.


Asunto(s)
Antiinflamatorios/farmacología , Bifidobacterium/efectos de los fármacos , Polisacáridos Fúngicos/farmacología , Inflamación/tratamiento farmacológico , Anaerobiosis/efectos de los fármacos , Antiinflamatorios/química , Fermentación/efectos de los fármacos , Polisacáridos Fúngicos/química , Humanos , Inflamación/microbiología , Inflamación/patología , Peso Molecular , Micelio/química
16.
Cancer Sci ; 112(10): 4050-4063, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34289209

RESUMEN

Astragalus polysaccharides (APS), the main effective component of Astragalus membranaceus, can inhibit tumor growth, but the underlying mechanisms remain unclear. Previous studies have suggested that APS can regulate the gut microenvironment, including the gut microbiota and fecal metabolites. In this work, our results showed that APS could control tumor growth in melanoma-bearing mice. It could reduce the number of myeloid-derived suppressor cells (MDSC), as well as the expression of MDSC-related molecule Arg-1 and cytokines IL-10 and TGF-ß, so that CD8+ T cells could kill tumor cells more effectively. However, while APS were administered with an antibiotic cocktail (ABX), MDSC could not be reduced, and the growth rate of tumors was accelerated. Consistent with the changes in MDSC, the serum levels of IL-6 and IL-1ß were lowest in the APS group. Meanwhile, we found that fecal suspension from mice in the APS group could also reduce the number of MDSC in tumor tissues. These results revealed that APS regulated the immune function in tumor-bearing mice through remodeling the gut microbiota. Next, we focused on the results of 16S rRNA, which showed that APS significantly regulated most microorganisms, such as Bifidobacterium pseudolongum, Lactobacillus johnsonii and Lactobacillus. According to the Spearman analysis, the changes in abundance of these microorganisms were related to the increase of metabolites like glutamate and creatine, which could control tumor growth. The present study demonstrates that APS attenuate the immunosuppressive activity of MDSC in melanoma-bearing mice by remodeling the gut microbiota and fecal metabolites. Our findings reveal the therapeutic potential of APS to control tumor growth.


Asunto(s)
Planta del Astrágalo/química , Linfocitos T CD8-positivos/inmunología , Microbioma Gastrointestinal/efectos de los fármacos , Melanoma/tratamiento farmacológico , Células Supresoras de Origen Mieloide/efectos de los fármacos , Polisacáridos/farmacología , Animales , Antibacterianos/administración & dosificación , Arginasa/efectos de los fármacos , Arginasa/metabolismo , Bifidobacterium/efectos de los fármacos , Bifidobacterium/metabolismo , Combinación de Medicamentos , Trasplante de Microbiota Fecal , Heces/microbiología , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Tolerancia Inmunológica , Interleucina-10/metabolismo , Interleucina-1beta/sangre , Interleucina-6/sangre , Lactobacillus/efectos de los fármacos , Masculino , Melanoma/inmunología , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , ARN Ribosómico 16S/análisis , Factor de Crecimiento Transformador beta/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/inmunología
17.
Sci Rep ; 11(1): 11264, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-34050197

RESUMEN

Short-term changes in dietary intake can induce changes in gut microbiome. While various dietary polyphenols have been shown to modulate gut microflora, the acute influence of polyphenol-rich mixed spices has not been explored in a controlled setting. We investigated the effects of a single serving of mixed spices Indian curry consumption, in two separate doses, on the gut microbiome in 15 healthy, Singaporean Chinese males, with age and BMI of 23.5 ± 2.4 years and 22.9 ± 2.2 kg/m2 respectively. We found that a low-polyphenol, no spices Dose 0 Control (D0C) meal led to an increase in Bacteroides and a decrease in Bifidobacterium. In comparison to D0C, there was significant suppression of Bacteroides (p < 0.05) and an increase in Bifidobacterium (p < 0.05) with increasing doses of curry meal Dose 1 Curry (D1C) and Dose 2 Curry (D2C) containing 6 g and 12 g mixed spices respectively. Significant correlations were also found between bacterial changes and plasma phenolic acids. No differences between treatments were observed in the alpha-diversity of the gut microflora. This study has shown that a single serving of mixed spices can significantly modify/restore certain commensal microbes, particularly in people who do not regularly consume these spices.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Polifenoles/farmacología , Bacteroides/efectos de los fármacos , Bifidobacterium/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Humanos , Masculino , Comidas , Periodo Posprandial/efectos de los fármacos , Singapur , Especias/microbiología , Adulto Joven
18.
Commun Biol ; 4(1): 541, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33972677

RESUMEN

This study aims to understand the mechanistic basis underlying the response of Bifidobacterium to lactulose ingestion in guts of healthy Japanese subjects, with specific focus on a lactulose transporter. An in vitro assay using mutant strains of Bifidobacterium longum subsp. longum 105-A shows that a solute-binding protein with locus tag number BL105A_0502 (termed LT-SBP) is primarily involved in lactulose uptake. By quantifying faecal abundance of LT-SBP orthologues, which is defined by phylogenetic analysis, we find that subjects with 107 to 109 copies of the genes per gram of faeces before lactulose ingestion show a marked increase in Bifidobacterium after ingestion, suggesting the presence of thresholds between responders and non-responders to lactulose. These results help predict the prebiotics-responder and non-responder status and provide an insight into clinical interventions that test the efficacy of prebiotics.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Bacterianas/metabolismo , Bifidobacterium/crecimiento & desarrollo , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Lactulosa/administración & dosificación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Bifidobacterium/efectos de los fármacos , Bifidobacterium/metabolismo , Estudios Transversales , Femenino , Fármacos Gastrointestinales/administración & dosificación , Tracto Gastrointestinal/efectos de los fármacos , Humanos , Persona de Mediana Edad , Adulto Joven
19.
Nutrients ; 13(4)2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33920960

RESUMEN

ß-glucan consumption is known for its beneficial health effects, but the mode of action is unclear. While humans and mice lack the required enzymes to digest ß-glucans, certain intestinal microbes can digest ß-glucans, triggering gut microbial changes. Curdlan, a particulate ß-glucan isolated from Alcaligenes faecalis, is used as a food additive. In this study we determined the effect of curdlan intake in mice on the intestinal microbiota and dextran sodium sulfate (DSS)-induced intestinal inflammation. The effect of curdlan on the human intestinal microbiota was assessed using i-screen, an assay for studying anaerobic microbial interactions. Mice received oral gavage with vehicle or curdlan for 14 days followed by DSS for 7 days. The curdlan-fed group showed reduced weight loss and colonic inflammation compared to the vehicle-fed group. Curdlan intake did not induce general microbiota community changes, although a specific Bifidobacterium, closely related to Bifidobacterium choerinum, was observed to be 10- to 100-fold more prevalent in the curdlan-fed group under control and colitis conditions, respectively. When tested in i-screen, curdlan induced a global change in the microbial composition of the healthy intestinal microbiota from a human. Overall, these results suggest that dietary curdlan induces microbiota changes that could reduce intestinal inflammation.


Asunto(s)
Bifidobacterium/efectos de los fármacos , Colitis/tratamiento farmacológico , Dieta/métodos , Microbioma Gastrointestinal/efectos de los fármacos , beta-Glucanos/farmacología , Animales , Colitis/inducido químicamente , Colon/metabolismo , Sulfato de Dextran , Humanos , Ratones
20.
Genes (Basel) ; 12(4)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33924280

RESUMEN

Bifidobacterium has a diverse host range and shows several beneficial properties to the hosts. Many species should have co-evolved with their hosts, but the phylogeny of Bifidobacterium is dissimilar to that of host animals. The discrepancy could be linked to the niche-specific evolution due to hosts' dietary carbohydrates. We investigated the relationship between bifidobacteria and their host diet using a comparative genomics approach. Since carbohydrates are the main class of nutrients for bifidobacterial growth, we examined the distribution of carbohydrate-active enzymes, in particular glycoside hydrolases (GHs) that metabolize unique oligosaccharides. When bifidobacterial species are classified by their distribution of GH genes, five groups arose according to their hosts' feeding behavior. The distribution of GH genes was only weakly associated with the phylogeny of the host animals or with genomic features such as genome size. Thus, the hosts' dietary pattern is the key determinant of the distribution and evolution of GH genes.


Asunto(s)
Bifidobacterium/genética , Carbohidratos de la Dieta/farmacología , Glicósido Hidrolasas/genética , Animales , Composición de Base , Bifidobacterium/clasificación , Bifidobacterium/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Tamaño del Genoma , Genoma Bacteriano , Glicósido Hidrolasas/efectos de los fármacos , Interacciones Huésped-Patógeno , Familia de Multigenes , Filogenia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA