Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 638
Filter
1.
Gut Microbes ; 16(1): 2387139, 2024.
Article in English | MEDLINE | ID: mdl-39106231

ABSTRACT

Bacteriocins are broad or narrow-spectrum antimicrobial compounds that have received significant scientific attention due to their potential to treat infections caused by antibiotic-resistant pathogenic bacteria. The genome of Bifidobacterium pseudocatenulatum MM0196, an antimicrobial-producing, fecal isolate from a healthy pregnant woman, was shown to contain a gene cluster predicted to encode Pseudocin 196, a novel lantibiotic, in addition to proteins involved in its processing, transport and immunity. Following antimicrobial assessment against various indicator strains, protease-sensitive Pseudocin 196 was purified to homogeneity from cell-free supernatant. MALDI TOF mass spectrometry confirmed that the purified antimicrobial compound corresponds to a molecular mass of 2679 Da, which is consistent with that deduced from its genetic origin. Pseudocin 196 is classified as a lantibiotic based on its similarity to lacticin 481, a lanthionine ring-containing lantibiotic produced by Lactococcus lactis. Pseudocin 196, the first reported bacteriocin produced by a B. pseudocatenulatum species of human origin, was shown to inhibit clinically relevant pathogens, such as Clostridium spp. and Streptococcus spp. thereby highlighting the potential application of this strain as a probiotic to treat and prevent bacterial infections.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Bifidobacterium , Bacteriocins/pharmacology , Bacteriocins/genetics , Bacteriocins/metabolism , Bacteriocins/chemistry , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Bifidobacterium/genetics , Bifidobacterium/drug effects , Bifidobacterium/metabolism , Female , Clostridium/genetics , Clostridium/drug effects , Clostridium/metabolism , Feces/microbiology , Streptococcus/drug effects , Streptococcus/genetics , Streptococcus/metabolism , Pregnancy , Multigene Family , Microbial Sensitivity Tests , Genome, Bacterial , Probiotics/pharmacology
2.
Food Res Int ; 191: 114720, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39059916

ABSTRACT

Mangosteen (Garcinia mangostana L.) is a tasty, polyphenol-rich tropical fruit. The edible part is highly appreciated by its aroma, taste and texture. The non-edible part, rich in polyphenols, has been traditionally used in Thai medicine. In this work, flavonoids and phenolic acid/derivatives were identified in mangosteen extracts (ME) from edible and non-edible portions. We first studied the effects of MEs on the growth, metabolism, antioxidant capacity, biofilm formation and antimicrobial capacity of eight bifidobacteria and lactobacilli strains from intestinal origin and two commercial probiotic strains (BB536 and GG). ME concentrations higher than 10-20 % were inhibitory for all strains. However, ME concentrations of 5 % significantly (P < 0.01) increased all strains antioxidant capacity, reduced biofilm-formation, and enhanced inhibition against Gram-positive pathogens. To apply these knowledge, bifunctional fermented milk products were elaborated with 5 % ME and individual strains, which were selected taking into account their growth with ME, and the widest range of values on antioxidant capacity, biofilm formation and antimicrobial activity (bifidobacteria INIA P2 and INIA P467, lactobacilli INIA P459 and INIA P708, and reference strain GG). Most strains survived well manufacture, refrigerated storage and an in vitro simulation of major conditions encountered in the gastrointestinal tract. As expected, products supplemented with ME showed higher polyphenol content and antioxidant capacity levels than control. After sensory evaluation, products containing strains INIA P2, INIA P708 and GG outstood as best.


Subject(s)
Antioxidants , Biofilms , Cultured Milk Products , Garcinia mangostana , Lactobacillus , Plant Extracts , Plant Extracts/pharmacology , Garcinia mangostana/chemistry , Biofilms/drug effects , Biofilms/growth & development , Antioxidants/pharmacology , Lactobacillus/drug effects , Lactobacillus/metabolism , Cultured Milk Products/microbiology , Bifidobacterium/drug effects , Bifidobacterium/growth & development , Bifidobacterium/metabolism , Probiotics , Flavonoids/pharmacology , Flavonoids/analysis , Humans , Fruit/chemistry , Fruit/microbiology , Fermentation , Hydroxybenzoates/pharmacology , Gastrointestinal Microbiome/drug effects , Polyphenols/pharmacology
3.
BMC Genomics ; 25(1): 718, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39054474

ABSTRACT

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.


Subject(s)
Bifidobacterium , Enterococcus , Gastrointestinal Microbiome , Humans , Zimbabwe , Infant , Gastrointestinal Microbiome/genetics , Enterococcus/genetics , Enterococcus/drug effects , Enterococcus/isolation & purification , Bifidobacterium/genetics , Bifidobacterium/isolation & purification , Bifidobacterium/drug effects , Genomics , Genome, Bacterial , Feces/microbiology , Whole Genome Sequencing , Cohort Studies , Phylogeny
4.
Nutrients ; 16(13)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38999750

ABSTRACT

(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.


Subject(s)
Cholesterol , Feces , Fermentation , Gastrointestinal Microbiome , Oligosaccharides , Pectins , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Pectins/pharmacology , Pectins/metabolism , Cholesterol/metabolism , Oligosaccharides/pharmacology , Feces/microbiology , Humans , Prebiotics , Male , Metabolomics , Fatty Acids, Volatile/metabolism , Bifidobacterium/metabolism , Bifidobacterium/drug effects , Female , Bacteria/metabolism , Bacteria/drug effects , Bacteria/classification , Citrus
5.
Int J Biol Macromol ; 273(Pt 1): 133053, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38857723

ABSTRACT

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.


Subject(s)
Fermentation , Gastrointestinal Microbiome , Oligosaccharides , Trehalose , Trehalose/pharmacology , Trehalose/chemistry , Gastrointestinal Microbiome/drug effects , Humans , Oligosaccharides/pharmacology , Oligosaccharides/chemistry , Fermentation/drug effects , Feces/microbiology , Prebiotics , Bifidobacterium/drug effects , Bifidobacterium/metabolism
6.
Nutrients ; 16(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38892631

ABSTRACT

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.


Subject(s)
Antioxidants , Astragalus Plant , Diabetes Mellitus, Type 2 , Feces , Fermentation , Gastrointestinal Microbiome , Polysaccharides , Gastrointestinal Microbiome/drug effects , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Polysaccharides/pharmacology , Astragalus Plant/chemistry , Feces/microbiology , Antioxidants/pharmacology , Male , Female , Middle Aged , Thiamine/pharmacology , Thiamine/metabolism , Bifidobacterium/metabolism , Bifidobacterium/drug effects , Lactobacillus/metabolism , Lactobacillus/drug effects , Hypoglycemic Agents/pharmacology
7.
Gut Microbes ; 16(1): 2357176, 2024.
Article in English | MEDLINE | ID: mdl-38798019

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents , Bifidobacterium , Drug Resistance, Bacterial , Feces , Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Humans , Bifidobacterium/genetics , Bifidobacterium/drug effects , Bifidobacterium/isolation & purification , Infant, Newborn , Gastrointestinal Microbiome/drug effects , Feces/microbiology , Anti-Bacterial Agents/pharmacology , Female , RNA, Ribosomal, 16S/genetics , Drug Resistance, Bacterial/genetics , Male , Infant
8.
Biomed Pharmacother ; 174: 116561, 2024 May.
Article in English | MEDLINE | ID: mdl-38593705

ABSTRACT

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.


Subject(s)
Dietary Supplements , Gastrointestinal Microbiome , Healthy Volunteers , Pectins , Humans , Pectins/administration & dosage , Pectins/pharmacology , Gastrointestinal Microbiome/drug effects , Male , Adult , Double-Blind Method , Female , Young Adult , Rhinovirus/drug effects , Middle Aged , Feces/microbiology , Bifidobacterium/drug effects
9.
Benef Microbes ; 14(4): 317-334, 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-38661345

ABSTRACT

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.


Subject(s)
Bifidobacterium , Feces , Fructans , Inulin , Prebiotics , RNA, Ribosomal, 16S , Inulin/pharmacology , Humans , Bifidobacterium/genetics , Bifidobacterium/drug effects , Feces/microbiology , Fructans/pharmacology , RNA, Ribosomal, 16S/genetics , Oryza , Female , Male , Gastrointestinal Microbiome/drug effects , Adult , Chocolate , Young Adult , In Situ Hybridization, Fluorescence
10.
Nutrients ; 14(2)2022 Jan 12.
Article in English | MEDLINE | ID: mdl-35057489

ABSTRACT

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.


Subject(s)
Constipation/microbiology , Defecation/drug effects , Feces/microbiology , Galactose/pharmacology , Oligosaccharides/pharmacology , Prebiotics/administration & dosage , Adolescent , Adult , Bifidobacterium/drug effects , Constipation/therapy , Double-Blind Method , Female , Gastrointestinal Microbiome/drug effects , Humans , Male , Middle Aged , Self Report , Young Adult
11.
Front Immunol ; 12: 772532, 2021.
Article in English | MEDLINE | ID: mdl-34970262

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/adverse effects , Gastrointestinal Microbiome/drug effects , Immune Checkpoint Inhibitors/administration & dosage , Lacticaseibacillus rhamnosus , Neoplasms/therapy , Probiotics/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Bacteroides/drug effects , Bacteroides/growth & development , Bifidobacterium/drug effects , Bifidobacterium/growth & development , Cell Line, Tumor , Feces/microbiology , Mice, Inbred BALB C , Neoplasms/microbiology
12.
Nutrients ; 13(11)2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34836092

ABSTRACT

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.


Subject(s)
Dietary Supplements , Gastrointestinal Microbiome/drug effects , Irritable Bowel Syndrome/drug therapy , Milk, Human/chemistry , Oligosaccharides/pharmacology , Adolescent , Adult , Aged , Bifidobacterium/drug effects , Double-Blind Method , Feces/microbiology , Female , Humans , Intestinal Mucosa/drug effects , Irritable Bowel Syndrome/microbiology , Male , Middle Aged , Treatment Outcome , Trisaccharides/pharmacology , Young Adult
13.
Nutrients ; 13(11)2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34836175

ABSTRACT

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.


Subject(s)
Kidney Diseases/drug therapy , Kidney/injuries , Lactobacillus plantarum , Probiotics/pharmacology , Sodium Chloride, Dietary/adverse effects , Animals , Bifidobacterium/drug effects , Diet/adverse effects , Gastrointestinal Microbiome/drug effects , Inflammation/etiology , Inflammation/metabolism , Intestines/metabolism , Kidney Diseases/etiology , Kidney Diseases/metabolism , Lactobacillus/drug effects , Male , Mice , Mice, Inbred C57BL , Prevotella/drug effects , RNA, Ribosomal, 16S/metabolism
14.
Nutrients ; 13(11)2021 Nov 06.
Article in English | MEDLINE | ID: mdl-34836220

ABSTRACT

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.


Subject(s)
Bile Acids and Salts/metabolism , Cholesterol/metabolism , Gastrointestinal Microbiome/drug effects , Plant Extracts/administration & dosage , Vitis/chemistry , Adult , Akkermansia/drug effects , Bifidobacterium/drug effects , Cholesterol/blood , Female , Healthy Volunteers , Humans , Male , Middle Aged , Pilot Projects , Polyphenols/metabolism , Powders , Triglycerides/blood , Verrucomicrobia/drug effects , Young Adult
15.
FASEB J ; 35(11): e21977, 2021 11.
Article in English | MEDLINE | ID: mdl-34613640

ABSTRACT

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.


Subject(s)
Bifidobacterium/drug effects , Inflammation/drug therapy , Probiotics , Salmonella Infections/drug therapy , Salmonella typhimurium/drug effects , Xylose , Animals , Gastrointestinal Microbiome/drug effects , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Probiotics/pharmacology , Probiotics/therapeutic use , Xylose/analogs & derivatives , Xylose/pharmacology , Xylose/therapeutic use
16.
Carbohydr Polym ; 273: 118546, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34560958

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Mannans/pharmacology , Amorphophallus/chemistry , Anti-Bacterial Agents/adverse effects , Bifidobacterium/drug effects , Enterobacteriaceae/drug effects , Fatty Acids, Volatile/metabolism , Fermentation , Humans , Hydrolysis , Mannans/chemistry , Molecular Structure , Molecular Weight , Particle Size , Trifluoroacetic Acid/chemistry , Viscosity
17.
Int J Biol Macromol ; 188: 586-594, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34403670

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bifidobacterium/drug effects , Fungal Polysaccharides/pharmacology , Inflammation/drug therapy , Anaerobiosis/drug effects , Anti-Inflammatory Agents/chemistry , Fermentation/drug effects , Fungal Polysaccharides/chemistry , Humans , Inflammation/microbiology , Inflammation/pathology , Molecular Weight , Mycelium/chemistry
18.
Cancer Sci ; 112(10): 4050-4063, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34289209

ABSTRACT

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.


Subject(s)
Astragalus Plant/chemistry , CD8-Positive T-Lymphocytes/immunology , Gastrointestinal Microbiome/drug effects , Melanoma/drug therapy , Myeloid-Derived Suppressor Cells/drug effects , Polysaccharides/pharmacology , Animals , Anti-Bacterial Agents/administration & dosage , Arginase/drug effects , Arginase/metabolism , Bifidobacterium/drug effects , Bifidobacterium/metabolism , Drug Combinations , Fecal Microbiota Transplantation , Feces/microbiology , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Immune Tolerance , Interleukin-10/metabolism , Interleukin-1beta/blood , Interleukin-6/blood , Lactobacillus/drug effects , Male , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , RNA, Ribosomal, 16S/analysis , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/immunology
19.
Commun Biol ; 4(1): 541, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33972677

ABSTRACT

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.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/metabolism , Bifidobacterium/growth & development , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/microbiology , Lactulose/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Bifidobacterium/drug effects , Bifidobacterium/metabolism , Cross-Sectional Studies , Female , Gastrointestinal Agents/administration & dosage , Gastrointestinal Tract/drug effects , Humans , Middle Aged , Young Adult
20.
Sci Rep ; 11(1): 11264, 2021 05 28.
Article in English | MEDLINE | ID: mdl-34050197

ABSTRACT

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.


Subject(s)
Gastrointestinal Microbiome/drug effects , Polyphenols/pharmacology , Bacteroides/drug effects , Bifidobacterium/drug effects , Eating/drug effects , Humans , Male , Meals , Postprandial Period/drug effects , Singapore , Spices/microbiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL