Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Benef Microbes ; 11(6): 527-534, 2020 Oct 12.
Article in English | MEDLINE | ID: mdl-33032471

ABSTRACT

Bifidobacterium infantis NLS super strain (B. infantis NLS-SS) was previously shown to alleviate gastrointestinal symptoms in newly diagnosed coeliac disease (CD) patients consuming gluten. A high proportion of patients following a gluten-free diet experiences symptoms despite dietary compliance. The role of B. infantis in persistently symptomatic CD patients has not been explored. The aim of the study was to evaluate the effect of B. infantis NLS-SS on persistent gastrointestinal symptoms in patients with CD following a long-term GFD. We conducted a randomised, cross-over, double-blind, placebo-controlled trial in symptomatic adult CD patients on a GFD for at least two years. After one-week run-in, patients were randomised to B. infantis NLS-SS or placebo for 3 weeks with cross-over after a 2-week wash-out period. We estimated changes (Δ) in celiac symptom index (CSI) before and after treatment. Stool samples were collected for faecal microbiota analysis (16S rRNA sequencing). Gluten immunogenic peptide (GIP) excretion in stool and urine samples was measured at each study period. Eighteen patients were enrolled; six patients were excluded due violations in protocol. For patients with the highest clinical burden, CD symptoms were lower in probiotic than in placebo treatment (P=0.046). B. infantis and placebo treated groups had different microbiota profiles as assessed by beta diversity clustering. In probiotic treated groups, we observed an increase in abundance of B. infantis. Treatment with B. infantis was associated with decreased abundance of Ruminococcus sp. and Bifidobacterium adolescentis. GIP excretion in stools and urine was similar at each treatment period. There were no differences in adverse effects between the two groups. B. infantis NLS-SS improves specific CD symptoms in a subset of highly symptomatic treated patients (GFD). This is associated with a shift in stool microbiota profile. Larger studies are needed to confirm these findings. ClinicalTrials.gov: NCT03271138.


Subject(s)
Bifidobacterium longum subspecies infantis , Celiac Disease/therapy , Diet, Gluten-Free , Gastrointestinal Microbiome , Probiotics/therapeutic use , Adult , Bacterial Load , Bifidobacterium longum subspecies infantis/growth & development , Celiac Disease/diet therapy , Celiac Disease/microbiology , Cross-Over Studies , Double-Blind Method , Feces/chemistry , Feces/microbiology , Female , Glutens/analysis , Glutens/urine , Humans , Male , Middle Aged , Peptide Fragments/analysis , Peptide Fragments/urine , Ruminococcus/growth & development
2.
J Food Sci ; 85(5): 1596-1604, 2020 May.
Article in English | MEDLINE | ID: mdl-32267970

ABSTRACT

The human gut microbiome has been classified into three distinct enterotypes (Bacteroides, Prevotella, and Ruminococcus). The relationship between probiotics and gut enterotype is not yet clear. Cayenne pepper is effective in vitro as a prebiotic for Bifidobacteria and Lactobacilli, so cayenne ingestion with probiotics may lead to more profound gut microbial shifts. We aimed to determine whether probiotics (with or without cayenne pepper) alter gut bacterial community composition and if these changes are associated with the original gut enterotype of the individual. A total of 27 adult participants provided three fecal samples: prior to probiotic treatment (baseline), post probiotic treatment (probiotic), and post probiotic plus cayenne pepper treatment (probiotic + cayenne). DNA was extracted, amplified, and the V4 region sequenced on the Illumina MiSeq platform using V2 chemistry. Sequence reads were processed in mothur and assigned using the SILVA reference by phylotype. Three enterotypes characterized the study population-Bacteroides (B; n = 6), Prevotella (P; n = 11), and Ruminoccocus (R; n = 10). There was no significant increase in probiotic genera in fecal samples after treatment periods. Alpha diversity scores were significantly lower in B-type but not in P- or R-type individuals after probiotic treatment. For the majority of individuals, their enterotype remained constant regardless of probiotic (and cayenne) treatment. This suggests that baseline gut community characteristics and enterotype classification influence responsiveness to probiotic treatment, but that enterotype is stable across administration of prebiotic and probiotics. PRACTICAL APPLICATION: A person's gut microbial community influences their responsiveness to probiotics and prebiotic ingredients. Consumers must understand that it is difficult to shift their gut microbiota even with simultaneous administration of prebiotic and probiotic. Greater understanding of these phenomena will enable consumers to choose the most efficacious products for their needs.


Subject(s)
Bifidobacterium/physiology , Gastrointestinal Microbiome/drug effects , Lactobacillus/physiology , Probiotics/administration & dosage , Adult , Bacteroides/genetics , Bacteroides/growth & development , Bacteroides/isolation & purification , Feces/microbiology , Female , Humans , Male , Prebiotics/administration & dosage , Prevotella/genetics , Prevotella/growth & development , Prevotella/isolation & purification , Ruminococcus/genetics , Ruminococcus/growth & development , Ruminococcus/isolation & purification , Young Adult
3.
Gut Microbes ; 10(4): 439-446, 2019.
Article in English | MEDLINE | ID: mdl-31309868

ABSTRACT

Gut microbiome community dynamics are maintained by complex microbe-microbe and microbe-host interactions, which can be disturbed by stress. In vivo studies on the dynamics and manipulation of those interactions are costly and slow, but can be accelerated using in vitro fermentation. Herein, in vitro fermentation was used to determine how an acute stressor, a sudden change in diet, impacts inter-bacterial species competition for resistant starch-supplemented medium (RSM). Fermentation vessels were seeded with fecal samples collected from 10 individuals consuming a habitual diet or U.S. military rations for 21 days. Lactobacillus spp. growth in response to RSM was attenuated following ration consumption, whereas growth of Ruminococcus bromii was enhanced. These differences were not evident in the pre-fermentation samples. Findings demonstrate how incorporating in vitro fermentation into clinical studies can increase understanding of stress-induced changes in nutrient-microbiome dynamics, and suggest that sudden changes in diet may impact inter-species competition for substrates.


Subject(s)
Gastrointestinal Microbiome/drug effects , Starch/pharmacology , Adolescent , Adult , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Bacteria/metabolism , Culture Media/chemistry , DNA, Bacterial/genetics , Feces/microbiology , Fermentation , Gastrointestinal Microbiome/genetics , Humans , Lactobacillus/genetics , Lactobacillus/growth & development , Lactobacillus/metabolism , Male , Middle Aged , Military Personnel , RNA, Ribosomal, 16S/genetics , Ruminococcus/genetics , Ruminococcus/growth & development , Ruminococcus/metabolism , Starch/chemistry , Starch/metabolism , Young Adult
4.
Environ Pollut ; 248: 388-396, 2019 May.
Article in English | MEDLINE | ID: mdl-30825764

ABSTRACT

N-nitrosamines (NAs) are an emerging group of disinfection by-products that occur as a mixture in drinking water. Although the potency of the individual NA components in drinking water is negligible, their combined effect is rarely reported. We tested whether multicomponent NAs mixtures at environmentally relevant levels would produce significant effects when each component was combined at extremely low concentrations i.e. a million times lower than its No Observed Effect Concentration (NOEC). Mixture L (the maximum values detected in drinking water) or mixture M (one order of magnitude higher than detected) were fed to male and female Sprague-Dawley (SD) rats since PND 28 for seven days. We found that the body weight gains and the triglyceride (TG) levels increased significantly in mixture M treated male rats. Correspondingly, an obesogenic microbiota profile was obtained in the mixture M treated young male rat: Firmicutes/Bacteroidetes and the obesity-related taxa including Alistipes, Ruminococcus were enriched. Collectively, this is the first in vivo demonstration of NAs mixtures at environmentally relevant levels. Despite the complicated relationship between gut microbiota and obesity, our study has demonstrated that changes in gut microbiota may contribute to the development of obesity after the exposure. Our results highlight that changes in gut microbiota could be a risk factor for obesity, which emphasizes the need to include gut microbiota in the traditional mammalian risk assessment.


Subject(s)
Drinking Water/chemistry , Gastrointestinal Microbiome/drug effects , Nitrosamines/adverse effects , Obesity/chemically induced , Triglycerides/blood , Water Pollutants, Chemical/adverse effects , Weight Gain/drug effects , Animals , Bacteroidetes/growth & development , Disinfection , Female , Firmicutes/growth & development , Male , Rats , Rats, Sprague-Dawley , Ruminococcus/growth & development , Water Purification/methods
5.
PLoS One ; 14(1): e0199274, 2019.
Article in English | MEDLINE | ID: mdl-30699108

ABSTRACT

BACKGROUND: Resistant starch is a prebiotic metabolized by the gut bacteria. It has been shown to attenuate chronic kidney disease (CKD) progression in rats. Previous studies employed taxonomic analysis using 16S rRNA sequencing and untargeted metabolomics profiling. Here we expand these studies by metaproteomics, gaining new insight into the host-microbiome interaction. METHODS: Differences between cecum contents in CKD rats fed a diet containing resistant starch with those fed a diet containing digestible starch were examined by comparative metaproteomics analysis. Taxonomic information was obtained using unique protein sequences. Our methodology results in quantitative data covering both host and bacterial proteins. RESULTS: 5,834 proteins were quantified, with 947 proteins originating from the host organism. Taxonomic information derived from metaproteomics data surpassed previous 16S RNA analysis, and reached species resolutions for moderately abundant taxonomic groups. In particular, the Ruminococcaceae family becomes well resolved-with butyrate producers and amylolytic species such as R. bromii clearly visible and significantly higher while fibrolytic species such as R. flavefaciens are significantly lower with resistant starch feeding. The observed changes in protein patterns are consistent with fiber-associated improvement in CKD phenotype. Several known host CKD-associated proteins and biomarkers of impaired kidney function were significantly reduced with resistant starch supplementation. Data are available via ProteomeXchange with identifier PXD008845. CONCLUSIONS: Metaproteomics analysis of cecum contents of CKD rats with and without resistant starch supplementation reveals changes within gut microbiota at unprecedented resolution, providing both functional and taxonomic information. Proteins and organisms differentially abundant with RS supplementation point toward a shift from mucin degraders to butyrate producers.


Subject(s)
Bacterial Proteins/analysis , Cecum/microbiology , Gastrointestinal Microbiome , Proteome/analysis , Proteomics , Renal Insufficiency, Chronic/chemically induced , Ruminococcus , Starch/adverse effects , Animals , Disease Progression , Male , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/microbiology , Ruminococcus/classification , Ruminococcus/growth & development , Starch/pharmacology
6.
Int J Mol Sci ; 19(12)2018 Dec 17.
Article in English | MEDLINE | ID: mdl-30562947

ABSTRACT

Intestinal microbiota and their metabolites are strongly associated with host physiology. Developments in DNA sequencing and mass spectrometry technologies have allowed us to obtain additional data that enhance our understanding of the interactions among microbiota, metabolites, and the host. However, the strategies used to analyze these datasets are not yet well developed. Here, we describe an original analytical strategy, metabologenomics, consisting of an integrated analysis of mass spectrometry-based metabolome data and high-throughput-sequencing-based microbiome data. Using this approach, we compared data obtained from C57BL/6J mice fed an American diet (AD), which contained higher amounts of fat and fiber, to those from mice fed control rodent diet. The feces of the AD mice contained higher amounts of butyrate and propionate, and higher relative abundances of Oscillospira and Ruminococcus. The amount of butyrate positively correlated with the abundance of these bacterial genera. Furthermore, integrated analysis of the metabolome data and the predicted metagenomic data from Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) indicated that the abundance of genes associated with butyrate metabolism positively correlated with butyrate amounts. Thus, our metabologenomic approach is expected to provide new insights and understanding of intestinal metabolic dynamics in complex microbial ecosystems.


Subject(s)
Diet , Gastrointestinal Microbiome , Metabolome , Metagenomics , Ruminococcus , Animals , Humans , Male , Mice , Ruminococcus/genetics , Ruminococcus/growth & development
7.
J Anim Sci ; 96(10): 4360-4367, 2018 09 29.
Article in English | MEDLINE | ID: mdl-30060161

ABSTRACT

Animal science nutrition studies are increasingly focusing on finding solutions to reduce methane (CH4) emissions. In the present study, we evaluated the effect of reductive acetogenic bacteria [acetogen probiotics (AP)] and lauric acid (LA) on in vivo rumen fermentation and microbial populations in Hanwoo steers. Four cannulated Hanwoo steers (392 ± 14 kg) were analyzed in a 4 × 4 Latin square design and were placed in hood-type chambers. They were fed similar amounts of concentrate and rice straw within and experimental design as follows: control (Con; 40 g DM basal feed, nonaddition of AP or LA), T1 = LA (40 g DM basal feed mixed with 40 g LA), T2 = AP (40 g DM basal feed, fermented with AP), and T3 = LA + AP (40 g DM basal feed, fermented with AP and mixed with 40 g LA). The animals were acclimatized to the diet for 15 d, followed by 6 d of the experimental period. Rumen fluid samples for metabolite and molecular analyses were collected 6 h after the morning feeding, with 2-h collection intervals. The enteric CH4 production was monitored on the last 2 d of the experimental period. Concentrations of total volatile fatty acids increased with the increase in time after feeding. Acetate, propionate, and butyrate concentrations were observed to be higher in the treatments than in Con. The addition of LA and AP reduced CH4 emission compared with that of Con (P < 0.01). Nuclear magnetic resonance spectroscopy results revealed no correlation between the LA and Con groups, but AP showed a correlation with LA and Con. Reduction in the number of protozoa which was accompanied by a decrease, because methanogens live symbiotically with protozoa. Supplementation of AP or LA alone and in combination decreased (P < 0.05) the methanogen population, whereas supplementation of LA alone significantly increased (P < 0.05) Ruminococcus flavefaciens and slightly increased total fungi. Thus, dietary supplementation of LA and AP has inhibitory effects on CH4 production in Hanwoo cattle. If the effects of this method can be maintained, reductive acetogens could become an important part of strategies to lower ruminant CH4 emissions.


Subject(s)
Acetates/metabolism , Bacteria/metabolism , Cattle/physiology , Lauric Acids/pharmacology , Methane/metabolism , Probiotics/pharmacology , Animal Feed/analysis , Animals , Bacteria/isolation & purification , Cattle/microbiology , Diet/veterinary , Fatty Acids, Volatile/metabolism , Fermentation/drug effects , Fungi/growth & development , Male , Republic of Korea , Rumen/metabolism , Ruminococcus/growth & development
8.
Twin Res Hum Genet ; 21(3): 203-213, 2018 06.
Article in English | MEDLINE | ID: mdl-29792248

ABSTRACT

OBJECTIVE: The human gut microbiota has been demonstrated to be associated with a number of host phenotypes, including obesity and a number of obesity-associated phenotypes. This study is aimed at further understanding and describing the relationship between the gut microbiota and obesity-associated measurements obtained from human participants. SUBJECTS/METHODS: Here, we utilize genetically informative study designs, including a four-corners design (extremes of genetic risk for BMI and of observed BMI; N = 50) and the BMI monozygotic (MZ) discordant twin pair design (N = 30), in order to help delineate the role of host genetics and the gut microbiota in the development of obesity. RESULTS: Our results highlight a negative association between BMI and alpha diversity of the gut microbiota. The low genetic risk/high BMI group of individuals had a lower gut microbiota alpha diversity when compared to the other three groups. Although the difference in alpha diversity between the lean and heavy groups of the BMI-discordant MZ twin design did not achieve significance, this difference was observed to be in the expected direction, with the heavier participants having a lower average alpha diversity. We have also identified nine OTUs observed to be associated with either a leaner or heavier phenotype, with enrichment for OTUs classified to the Ruminococcaceae and Oxalobacteraceae taxonomic families. CONCLUSION: Our study presents evidence of a relationship between BMI and alpha diversity of the gut microbiota. In addition to these findings, a number of OTUs were found to be significantly associated with host BMI. These findings may highlight separate subtypes of obesity, one driven by genetic factors, the other more heavily influenced by environmental factors.


Subject(s)
Body Mass Index , Gastrointestinal Microbiome , Oxalobacteraceae/classification , Ruminococcus/classification , Twins, Monozygotic , Adult , Female , Humans , Male , Oxalobacteraceae/growth & development , Ruminococcus/growth & development
9.
J Nutr Biochem ; 49: 89-100, 2017 11.
Article in English | MEDLINE | ID: mdl-28915390

ABSTRACT

Common beans (Phaseolus vulgaris L.) are enriched in non-digestible fermentable carbohydrates and phenolic compounds that can modulate the colonic microenvironment (microbiota and host epithelial barrier) to improve gut health. In a comprehensive assessment of the impact of two commonly consumed bean varieties (differing in levels and types of phenolic compounds) within the colonic microenvironment, C57Bl/6 mice were fed diets supplemented with 20% cooked navy bean (NB) or black bean (BB) flours or an isocaloric basal diet control (BD) for 3 weeks. NB and BB similarly altered the fecal microbiota community structure (16S rRNA sequencing) notably by increasing the abundance of carbohydrate fermenting bacteria such as Prevotella, S24-7 and Ruminococcus flavefaciens, which coincided with enhanced short chain fatty acid (SCFA) production (microbial-derived carbohydrate fermentation products) and colonic expression of the SCFA receptors GPR-41/-43/-109a. Both NB and BB enhanced multiple aspects of mucus and epithelial barrier integrity vs. BD including: (i) goblet cell number, crypt mucus content and mucin mRNA expression, (ii) anti-microbial defenses (Reg3γ), (iii) crypt length and epithelial cell proliferation, (iv) apical junctional complex components (occludin, JAM-A, ZO-1 and E-cadherin) mRNA expression and (v) reduced serum endotoxin concentrations. Interestingly, biomarkers of colon barrier integrity (crypt height, mucus content, cell proliferation and goblet cell number) were enhanced in BB vs. NB-fed mice, suggesting added benefits attributable to unique BB components (e.g., phenolics). Overall, NB and BB improved baseline colonic microenvironment function by altering the microbial community structure and activity and promoting colon barrier integrity and function; effects which may prove beneficial in attenuating gut-associated diseases.


Subject(s)
Colon/metabolism , Dysbiosis/prevention & control , Functional Food , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/metabolism , Phytohemagglutinins , Seeds , Animals , Biomarkers/metabolism , Cell Proliferation , Cellular Microenvironment , Colon/cytology , Colon/microbiology , Colon/pathology , Dietary Carbohydrates/administration & dosage , Dietary Carbohydrates/metabolism , Dietary Fiber/administration & dosage , Dietary Fiber/metabolism , Dysbiosis/metabolism , Dysbiosis/microbiology , Dysbiosis/pathology , Feces/microbiology , Fermentation , Gene Expression Regulation, Developmental , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice, Inbred C57BL , Molecular Typing , Prevotella/classification , Prevotella/growth & development , Prevotella/isolation & purification , Random Allocation , Ruminococcus/chemistry , Ruminococcus/growth & development , Ruminococcus/isolation & purification
10.
Biomed Res Int ; 2017: 4969076, 2017.
Article in English | MEDLINE | ID: mdl-29457028

ABSTRACT

Ruminants derived products have a prominent role in diets and economy worldwide; therefore, the capability to control the rumen microbial ecosystem, for ameliorating their quality, is of fundamental importance in the livestock sector. The aim of this study was to evaluate the effect of dietary supplementation with chestnut and quebracho tannins on microbial community and fatty acid profile, in the rumen fluid of dairy ewes. Multivariate analysis of PCR-DGGE profiles of rumen microbial communities showed a correlation among the presence of chestnut or quebracho in the diet, the specific Butyrivibrio group DGGE profiles, the increase in 18:3 cis9, cis12, and cis15; 18:2 cis9 and cis12; 18:2 cis9 and trans11; 18:2 trans11 and cis15; and 18:1 trans11 content, and the decrease in 18:0 concentration. Phylogenetic analysis of DGGE band sequences revealed the presence of bacteria representatives related to the genera Hungatella, Ruminococcus, and Eubacterium and unclassified Lachnospiraceae family members, suggesting that these taxa could be affected by tannins presence in the diets. The results of this study showed that tannins from chestnut and quebracho can reduce the biohydrogenation of unsaturated fatty acids through changes in rumen microbial communities.


Subject(s)
Dietary Supplements , Rumen/microbiology , Tannins/administration & dosage , Aesculus/chemistry , Animal Feed , Animals , Digestion/drug effects , Eubacterium/drug effects , Eubacterium/genetics , Eubacterium/growth & development , Fatty Acids, Unsaturated , Female , Lactation/drug effects , Phylogeny , Rumen/drug effects , Ruminococcus/drug effects , Ruminococcus/genetics , Ruminococcus/growth & development , Sheep , Tannins/chemistry
12.
Anaerobe ; 42: 130-141, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27742572

ABSTRACT

Quercetin is one of the most abundant polyphenols found in fruits and vegetables. The ability of the gut microbiota to metabolize quercetin has been previously documented; however, the effect that quercetin may have on commensal gut microbes remains unclear. In the present study, the effects of quercetin on the commensal gut microbes Ruminococcus gauvreauii, Bifidobacterium catenulatum and Enterococcus caccae were determined through evaluation of growth patterns and cell morphology, and analysis of genetic expression profiles between quercetin treated and non-treated groups using Single Molecule RNA sequencing via Helicos technology. Results of this study revealed that phenotypically, quercetin did not prevent growth of Ruminococcus gauvreauii, mildly suppressed growth of Bifidobacterium catenulatum, and moderately inhibited growth of Enterococcus caccae. Genetic analysis revealed that in response to quercetin, Ruminococcus gauvreauii down regulated genes responsible for protein folding, purine synthesis and metabolism. Bifidobacterium catenulatum increased expression of the ABC transport pathway and decreased metabolic pathways and cell wall synthesis. Enterococcus caccae upregulated genes responsible for energy production and metabolism, and downregulated pathways of stress response, translation and sugar transport. For the first time, the effect of quercetin on the growth and genetic expression of three different commensal gut bacteria was documented. The data provides insight into the interactions between genetic regulation and growth. This is also a unique demonstration of how RNA single molecule sequencing can be used to study the gut microbiota.


Subject(s)
Bifidobacterium/drug effects , Enterococcus/drug effects , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation, Bacterial/drug effects , Polyphenols/pharmacology , Quercetin/pharmacology , Ruminococcus/drug effects , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Bifidobacterium/growth & development , Bifidobacterium/ultrastructure , Cell Wall/drug effects , Cell Wall/metabolism , Enterococcus/growth & development , Enterococcus/ultrastructure , Gastrointestinal Microbiome/physiology , Gene Expression Profiling , Humans , Metabolic Networks and Pathways/drug effects , Molecular Sequence Annotation , Protein Folding/drug effects , Purines/biosynthesis , Ruminococcus/growth & development , Ruminococcus/ultrastructure , Sequence Analysis, RNA , Symbiosis
13.
Microbiome ; 4(1): 53, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27717398

ABSTRACT

BACKGROUND: Clostridium difficile is the most common known cause of antibiotic-associated diarrhea. Upon the disturbance of gut microbiota by antibiotics, C. difficile establishes growth and releases toxins A and B, which cause tissue damage in the host. The symptoms of C. difficile infection disease range from mild diarrhea to pseudomembranous colitis and toxic megacolon. Interestingly, 10-50 % of infants are asymptomatic carriers of C. difficile. This longitudinal study of the C. difficile colonization in an infant revealed the dynamics of C. difficile presence in gut microbiota. METHODS: Fifty fecal samples, collected weekly between 5.5 and 17 months of age from a female infant who was an asymptomatic carrier of C. difficile, were analyzed by 16S rRNA gene sequencing. RESULTS: Colonization switching between toxigenic and non-toxigenic C. difficile strains as well as more than 100,000-fold fluctuations of C. difficile counts were observed. C. difficile toxins were detected during the testing period in some infant stool samples, but the infant never had diarrhea. Although fecal microbiota was stable during breast feeding, a dramatic and permanent change of microbiota composition was observed within 5 days of the transition from human milk to cow milk. A rapid decline and eventual disappearance of C. difficile coincided with weaning at 12.5 months. An increase in the relative abundance of Bacteroides spp., Blautia spp., Parabacteroides spp., Coprococcus spp., Ruminococcus spp., and Oscillospira spp. and a decrease of Bifidobacterium spp., Lactobacillus spp., Escherichia spp., and Clostridium spp. were observed during weaning. The change in microbiome composition was accompanied by a gradual increase of fecal pH from 5.5 to 7. CONCLUSIONS: The bacterial groups that are less abundant in early infancy, and that increase in relative abundance after weaning, likely are responsible for the expulsion of C. difficile.


Subject(s)
Asymptomatic Infections , Bacterial Load , Breast Feeding , Clostridioides difficile/growth & development , Clostridium Infections/microbiology , Gastrointestinal Microbiome/physiology , Milk, Human , Weaning , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Bacteroides/growth & development , Bifidobacterium/growth & development , Clostridium/growth & development , Enterotoxins/metabolism , Escherichia/growth & development , Feces/microbiology , Female , Humans , Infant , Lactobacillus/growth & development , RNA, Ribosomal, 16S/genetics , Ruminococcus/growth & development
14.
Gut Microbes ; 7(4): 302-312, 2016 07 03.
Article in English | MEDLINE | ID: mdl-27223845

ABSTRACT

We previously identified and characterized an intramolecular trans-sialidase (IT-sialidase) in the gut symbiont Ruminococcus gnavus ATCC 29149, which is associated to the ability of the strain to grow on mucins. In this work we have obtained and analyzed the draft genome sequence of another R. gnavus mucin-degrader, ATCC 35913, isolated from a healthy individual. Transcriptomics analyses of both ATCC 29149 and ATCC 35913 strains confirmed that the strategy utilized by R. gnavus for mucin-degradation is focused on the utilization of terminal mucin glycans. R. gnavus ATCC 35913 also encodes a predicted IT-sialidase and harbors a Nan cluster dedicated to sialic acid utilization. We showed that the Nan cluster was upregulated when the strains were grown in presence of mucin. In addition we demonstrated that both R. gnavus strains were able to grow on 2,7-anyhydro-Neu5Ac, the IT-sialidase transglycosylation product, as a sole carbon source. Taken together these data further support the hypothesis that IT-sialidase expressing gut microbes, provide commensal bacteria such as R. gnavus with a nutritional competitive advantage, by accessing and transforming a source of nutrient to their own benefit.


Subject(s)
Bacterial Proteins/metabolism , Glycoproteins/metabolism , Mucins/metabolism , Neuraminidase/metabolism , Ruminococcus/metabolism , Bacterial Proteins/genetics , Genome, Bacterial , Glycoproteins/genetics , Neuraminidase/genetics , Ruminococcus/enzymology , Ruminococcus/genetics , Ruminococcus/growth & development
15.
Microb Ecol ; 72(4): 943-954, 2016 11.
Article in English | MEDLINE | ID: mdl-26984253

ABSTRACT

The mammalian gastrointestinal (GI) microbiome, which plays indispensable roles in host nutrition and health, is affected by numerous intrinsic and extrinsic factors. Among them, antibiotic (ATB) treatment is reported to have a significant effect on GI microbiome composition in humans and other animals. However, the impact of ATBs on the GI microbiome of free-ranging or even captive great apes remains poorly characterized. Here, we investigated the effect of cephalosporin treatment (delivered by intramuscular dart injection during a serious respiratory outbreak) on the GI microbiome of a wild habituated group of western lowland gorillas (Gorilla gorilla gorilla) in the Dzanga Sangha Protected Areas, Central African Republic. We examined 36 fecal samples from eight individuals, including samples before and after ATB treatment, and characterized the GI microbiome composition using Illumina-MiSeq sequencing of the bacterial 16S rRNA gene. The GI microbial profiles of samples from the same individuals before and after ATB administration indicate that the ATB treatment impacts GI microbiome stability and the relative abundance of particular bacterial taxa within the colonic ecosystem of wild gorillas. We observed a statistically significant increase in Firmicutes and a decrease in Bacteroidetes levels after ATB treatment. We found disruption of the fibrolytic community linked with a decrease of Ruminoccocus levels as a result of ATB treatment. Nevertheless, the nature of the changes observed after ATB treatment differs among gorillas and thus is dependent on the individual host. This study has important implications for ecology, management, and conservation of wild primates.


Subject(s)
Anti-Bacterial Agents/pharmacology , Ape Diseases/drug therapy , Cephalosporins/pharmacology , Gastrointestinal Microbiome/drug effects , Gorilla gorilla/microbiology , Animals , Bacteroidetes/growth & development , Central African Republic , Feces/microbiology , Firmicutes/growth & development , RNA, Ribosomal, 16S/genetics , Ruminococcus/growth & development
16.
J Sci Food Agric ; 96(13): 4565-74, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26910767

ABSTRACT

BACKGROUND: Condensed tannin (CT) fractions of different molecular weights (MWs) may affect rumen microbial metabolism by altering bacterial diversity. In this study the effects of unfractionated CTs (F0) and five CT fractions (F1-F5) of different MWs (F1, 1265.8 Da; F2, 1028.6 Da; F3, 652.2 Da; F4, 562.2 Da; F5, 469.6 Da) from Leucaena leucocephala hybrid-Rendang (LLR) on the structure and diversity of the rumen bacterial community were investigated in vitro. RESULTS: Real-time polymerase chain reaction assay showed that the total bacterial population was not significantly (P > 0.05) different among the dietary treatments. Inclusion of higher-MW CT fractions F1 and F2 significantly (P < 0.05) increased the Fibrobacter succinogenes population compared with F0 and CT fractions F3-F5. Although inclusion of F0 and CT fractions (F1-F5) significantly (P < 0.05) decreased the Ruminococcus flavefaciens population, there was no effect on the Ruminococcus albus population when compared with the control (without CTs). High-throughput sequencing of the V3 region of 16S rRNA showed that the relative abundance of genera Prevotella and unclassified Clostridiales was significantly (P < 0.05) decreased, corresponding with increasing MW of CT fractions, whereas cellulolytic bacteria of the genus Fibrobacter were significantly (P < 0.05) increased. Inclusion of higher-MW CT fractions F1 and/or F2 decreased the relative abundance of minor genera such as Ruminococcus, Streptococcus, Clostridium XIVa and Anaeroplasma but increased the relative abundance of Acinetobacter, Treponema, Selenomonas, Succiniclasticum and unclassified Spirochaetales compared with the control and lower-MW CT fractions. CONCLUSION: This study indicates that CT fractions of different MWs may play an important role in altering the structure and diversity of the rumen bacterial community in vitro, and the impact was more pronounced for CT fractions with higher MW. © 2016 Society of Chemical Industry.


Subject(s)
Diet/veterinary , Fabaceae/chemistry , Fibrobacter/growth & development , Gastrointestinal Contents/microbiology , Proanthocyanidins/administration & dosage , Rumen/microbiology , Ruminococcus/growth & development , Animals , Cattle , Clostridiales/classification , Clostridiales/growth & development , Clostridiales/isolation & purification , Clostridiales/metabolism , Crosses, Genetic , Digestion , Fibrobacter/classification , Fibrobacter/isolation & purification , Fibrobacter/metabolism , Gastrointestinal Microbiome , Male , Microbial Viability , Molecular Typing/veterinary , Molecular Weight , Plant Leaves/chemistry , Plant Shoots/chemistry , Prevotella/classification , Prevotella/growth & development , Prevotella/isolation & purification , Prevotella/metabolism , Proanthocyanidins/chemistry , Proanthocyanidins/isolation & purification , Proanthocyanidins/metabolism , Ruminococcus/classification , Ruminococcus/isolation & purification , Ruminococcus/metabolism , Species Specificity
17.
Water Res ; 93: 163-171, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26905795

ABSTRACT

Conversion of lactate to n-caproate had been described for the type strain Megasphaera elsdenii in batch systems. Recently, investigators have also described production of n-caproate from endogenous or exogenous lactate with batch-fed reactor microbiome systems. However, no reports exist of lactate to n-caproate conversion within a continuously fed bioreactor. Since continuously fed systems are advantageous for biotechnology production platforms, our objective was to develop such a system. Here, we demonstrated continuous lactate to n-caproate conversion for more than 165 days. The volumetric n-caproate production rate (productivity) was improved when we decreased the operating pH from 5.5 to 5.0, and was again improved when we utilized in-line product recovery via pertraction (membrane-based liquid-liquid extraction). We observed a maximum n-caproate productivity of 6.9 g COD/L-d for a period of 17 days at an L-lactate loading rate of 9.1 g COD/L-d, representing the highest sustained lactate to n-caproate conversion rate ever reported. We had to manage two competing lactate conversion pathways: 1) the reverse ß-oxidation pathway to n-caproate; and 2) the acrylate pathway to propionate. We found that maintaining a low residual lactate concentration in the bioreactor broth was necessary to direct lactate conversion towards n-caproate instead of propionate. These findings provide a foundation for the development of new resource recovery processes to produce higher-value liquid products (e.g., n-caproate) from carbon-rich wastewaters containing lactate or lactate precursors (e.g., carbohydrates).


Subject(s)
Bioreactors/microbiology , Caproates/metabolism , Lactic Acid/metabolism , Microbiota , Biomass , Clostridium/growth & development , Clostridium/metabolism , Hydrogen-Ion Concentration , Megasphaera elsdenii/growth & development , Megasphaera elsdenii/metabolism , Metabolic Networks and Pathways , Oxidation-Reduction , Ruminococcus/growth & development , Ruminococcus/metabolism , Time Factors
18.
Mucosal Immunol ; 9(1): 24-37, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25921339

ABSTRACT

HIV-1-associated disruption of intestinal homeostasis is a major factor contributing to chronic immune activation and inflammation. Dendritic cells (DCs) are crucial in maintaining intestinal homeostasis, but the impact of HIV-1 infection on intestinal DC number and function has not been extensively studied. We compared the frequency and activation/maturation status of colonic myeloid DC (mDC) subsets (CD1c(+) and CD1c(neg)) and plasmacytoid DCs in untreated HIV-1-infected subjects with uninfected controls. Colonic mDCs in HIV-1-infected subjects had increased CD40 but decreased CD83 expression, and CD40 expression on CD1c(+) mDCs positively correlated with mucosal HIV-1 viral load, with mucosal and systemic cytokine production, and with frequencies of activated colon and blood T cells. Percentage of CD83(+)CD1c(+) mDCs negatively correlated with frequencies of interferon-γ-producing colon CD4(+) and CD8(+) T cells. CD40 expression on CD1c(+) mDCs positively associated with abundance of high prevalence mucosal Prevotella copri and Prevotella stercorea but negatively associated with a number of low prevalence mucosal species, including Rumminococcus bromii. CD1c(+) mDC cytokine production was greater in response to in vitro stimulation with Prevotella species relative to R. bromii. These findings suggest that, during HIV infection, colonic mDCs become activated upon exposure to mucosal pathobiont bacteria leading to mucosal and systemic immune activation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Colon/immunology , Gastrointestinal Microbiome/immunology , HIV Infections/immunology , HIV-1/immunology , Mucous Membrane/immunology , Adult , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD1/genetics , Antigens, CD1/immunology , CD4-Positive T-Lymphocytes/microbiology , CD40 Antigens/genetics , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/microbiology , Case-Control Studies , Cell Lineage/immunology , Colon/microbiology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Female , Gene Expression Regulation , Glycoproteins/genetics , Glycoproteins/immunology , HIV Infections/microbiology , HIV Infections/pathology , Humans , Immunoglobulins/genetics , Immunoglobulins/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Activation , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Middle Aged , Mucous Membrane/microbiology , Prevotella/growth & development , Prevotella/immunology , Ruminococcus/growth & development , Ruminococcus/immunology , Signal Transduction , Viral Load , CD83 Antigen
19.
Microbiol Immunol ; 59(11): 643-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26486646

ABSTRACT

A large number of commensal bacteria inhabit the intestinal tract, and interbacterial communication among gut microbiota is thought to occur. In order to analyze symbiotic relationships between probiotic strains and the gut microbiota, a ring with a membrane filter fitted to the bottom was used for in vitro investigations. Test strains comprising probiotic nitto strains (Lactobacillus acidophilus NT and Bifidobacterium longum NT) and type strains (L. acidophilus JCM1132(T) and B. longum JCM1217(T) ) were obtained from diluted fecal samples using the membrane filter to simulate interbacterial communication. Bifidobacterium spp., Streptococcus pasteurianus, Collinsella aerofaciens, and Clostridium spp. were the most abundant gut bacteria detected before coculture with the test strains. Results of the coculture experiments indicated that the test strains significantly promote the growth of Ruminococcus gnavus, Ruminococcus torques, and Veillonella spp. and inhibit the growth of Sutterella wadsworthensis. Differences in the relative abundances of gut bacterial strains were furthermore observed after coculture of the fecal samples with each test strain. Bifidobacterium spp., which was detected as the dominant strain in the fecal samples, was found to be unaffected by coculture with the test strains. In the present study, interbacterial communication using bacterial metabolites between the test strains and the gut microbiota was demonstrated by the coculture technique. The detailed mechanisms and effects of the complex interbacterial communications that occur among the gut microbiota are, however, still unclear. Further investigation of these relationships by coculture of several fecal samples with probiotic strains is urgently required.


Subject(s)
Bifidobacterium/physiology , Clostridium/growth & development , Clostridium/physiology , Coculture Techniques/methods , Lactobacillus acidophilus/physiology , Ruminococcus/growth & development , Ruminococcus/physiology , Streptococcus/growth & development , Streptococcus/physiology , Veillonella/growth & development , Veillonella/physiology , Gastrointestinal Microbiome , Humans
20.
Mol Nutr Food Res ; 59(8): 1614-28, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25988339

ABSTRACT

SCOPE: Flaxseeds contain the phytoestrogens lignans that must be activated to enterolignans by intestinal bacteria. We investigated the impact of flaxseeds on fecal bacterial communities and their associations with fecal and blood metabolites. METHODS AND RESULTS: Nine healthy male adult subjects ingested 0.3 g/kg/day flaxseeds during 1 week. Gut bacteria as well as blood and fecal metabolites were analyzed. Ingestion of flaxseeds triggered a significant increase in the blood concentration of enterolignans, accompanied by fecal excretion of propionate and glycerol. Overall diversity and composition of dominant fecal bacteria remained individual specific throughout the study. Enterolactone production was linked to the abundance of two molecular species identified as Ruminococcus bromii and Ruminococcus lactaris. Most dominant species of the order Bacteroidales were positively associated with fecal concentrations of either acetic, isovaleric, or isobutyric acid, the latter being negatively correlated with blood levels of triglycerides. The relative sequence abundance of one Gemmiger species (Ruminococcaceae) and of Coprococcus comes (Lachnospiraceae) correlated positively with blood levels of LDL cholesterol and triglycerides, respectively. CONCLUSION: Flaxseeds increase enterolignan production but do not markedly alter fecal metabolome and dominant bacterial communities. The data underline the possible role of members of the family Ruminococcaceae in the regulation of enterolignan production and blood lipids.


Subject(s)
Diet , Flax/chemistry , Gastrointestinal Microbiome , Intestinal Mucosa/metabolism , Lignans/administration & dosage , Prebiotics , Seeds/chemistry , Adult , Bacteroidetes/classification , Bacteroidetes/growth & development , Bacteroidetes/isolation & purification , Bacteroidetes/metabolism , Biomarkers/analysis , Biomarkers/blood , Feces/chemistry , Feces/microbiology , Fermentation , Glycerol/analysis , Glycerol/metabolism , Humans , Intestinal Mucosa/microbiology , Lignans/analysis , Lignans/blood , Lignans/metabolism , Lipids/blood , Male , Molecular Typing , Pilot Projects , Propionates/analysis , Propionates/metabolism , Ruminococcus/classification , Ruminococcus/growth & development , Ruminococcus/isolation & purification , Ruminococcus/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...