Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Microorganisms ; 12(4)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38674741

ABSTRACT

Aging has been associated with a changed composition and function of the gut microbiota (GM). Here, we investigate the effects of the multi-strain probiotic HOWARU® Restore on GM composition and function in seniors. Ninety-eight healthy adult volunteers aged ≥75 years were enrolled in a randomised, double-blinded intervention (NCT02207140), where they received HOWARU Restore (1010 CFU) or the placebo daily for 24 weeks, with 45 volunteers from each group completing the intervention. Questionnaires monitoring the effects on gastro-intestinal discomfort and bowel movements were collected. Faecal samples for GM characterisation (qPCR, 16S rRNA gene amplicon sequencing) and metabolomics (GC-FID, 1H NMR) were collected at the baseline and after 24 weeks. In the probiotic group, self-reported gastro-intestinal discomfort in the form of flatulence was significantly decreased during the intervention. At the baseline, 151 'core species' (present in ≥95% of samples) were identified. Most core species belonged to the Lachnospiraceae and Ruminococcaceae families. Neither alpha diversity nor beta diversity or faecal metabolites was affected by probiotic intake. On the contrary, we observed high intra-individual GM stability, with 'individual' accounting for 72-75% of variation. In conclusion, 24 weeks of HOWARU Restore intake reduced gastro-intestinal discomfort in the form of flatulence in healthy seniors without significantly influencing GM composition or activity.

2.
Food Chem Toxicol ; 187: 114592, 2024 May.
Article in English | MEDLINE | ID: mdl-38493976

ABSTRACT

3-Fucosyllactose (3-FL) is one of the most abundant fucosylated oligosaccharides in human breast milk and is an approved infant formula ingredient world-wide. 3-FL functions as a prebiotic to promote early microbial colonization of the gut, increase pathogen resistance and modulate immune responses. To investigate safety and potential gut microbiota effects, 3-FL was fed for 21-days to farm piglets beginning on Postnatal Day (PND) 2. Fructooligosaccharide (FOS), an approved infant formula ingredient, was used as a reference control. Standard toxicological endpoints were evaluated, and the gut microbiota were assessed. Neither 3-FL (245.77 and 489.72 mg/kg/day for males and 246.57 and 494.18 mg/kg/day for females) nor FOS (489.44 and 496.33 mg/kg/day males and females, respectively) produced any adverse differences in growth, food intake or efficiency, clinical observations, or clinical or anatomic pathology changes. Differences in the gut microbiota after 3-FL consumption (versus control and FOS groups) included the absence of Bifidobacterium species from the piglets, enrichment of Prevotellamassilia timonensis, Blautia species, Mediterranea massiliensis, Lachnospiraceae incertae sedis, and Eubacterium coprostanoligens and lower relative abundance of Allisonella histaminiformans and Roseburia inulinivorans. This study further supports the safe use of 3-FL produced using biotechnology as a nutritional ingredient in foods.


Subject(s)
Infant Formula , Milk, Human , Infant , Male , Female , Humans , Animals , Swine , Trisaccharides/toxicity , Farms , Oligosaccharides/toxicity
3.
Nutrients ; 15(18)2023 Sep 07.
Article in English | MEDLINE | ID: mdl-37764689

ABSTRACT

As plant-based diets become more popular, there is an interest in developing innovations to improve the bioaccessibility of plant protein. In this study, seven probiotic strains (Bifidobacterium animalis subsp. lactis B420, B. lactis Bl-04, Lactobacillus acidophilus NCFM, Lacticaseibacillus rhamnosus HN001, Lacticaseibacillus paracasei subsp. paracasei Lpc-37, Lactiplantibacillus plantarum Lp-115, and Lactococcus lactis subsp. lactis Ll-23) were evaluated for their capacity to hydrolyze soy and pea protein ingredients in an in vitro digestion model of the upper gastrointestinal tract (UGIT). Compared to the control digestion of protein without a probiotic, all the studied strains were able to increase the digestion of soy or pea protein, as evidenced by an increase in free α-amino nitrogen (FAN) and/or free amino acid concentration. The increase in FAN varied between 13 and 33% depending on the protein substrate and probiotic strain. The survival of probiotic bacteria after exposure to digestive fluids was strain-dependent and may have affected the strain's capacity to function and aid in protein digestion in the gastrointestinal environment. Overall, our results from the standardized in vitro digestion model provide an approach to explore probiotics for improved plant protein digestion and bioaccessibility of amino acids; however, human clinical research is needed to evaluate the efficacy of probiotics on amino acid absorption and bioavailability in vivo.


Subject(s)
Bifidobacterium animalis , Lacticaseibacillus paracasei , Pea Proteins , Probiotics , Humans , Plant Proteins , Probiotics/metabolism , Lactobacillus acidophilus , Amino Acids
4.
Nutrients ; 15(16)2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37630731

ABSTRACT

Bifidobacterium animalis subsp. lactis HN019 is a probiotic with several documented human health benefits. Interest in probiotics has led to the development of new formats that probiotics, including HN019, can be supplemented into. In this study, we looked at common HN019 formats such as frozen culture and freeze-dried powder as well as supplementing it into the following food matrices: yogurts (dairy, soy, and oat based), xanthan gum-based tablets, pulpless orange juice, whey sports drink, and dark chocolate (70% cocoa). In this work, our aim was to investigate whether the food matrix that carried HN019 via simulated human digestion (a dual model system mimicking both upper and lower gastrointestinal digestion) influenced probiotic delivery. To that end, we validated and used a real-time qPCR assay to detect HN019 after simulated digestion. In addition, we also measured the effect on a panel of metabolites. After simulated digestion, we were able to detect HN019 from all the matrices tested, and the observed changes to the metabolite profile were consistent with those expected from the food matrix used. In conclusion, this work suggests that the food matrix supplemented with HN019 did not interfere with delivery to the colon via simulated human digestion.


Subject(s)
Bifidobacterium , Digestion , Humans , Bifidobacterium/genetics , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Lactic Acid/metabolism , Fatty Acids/metabolism , Colon/metabolism , Colon/microbiology
5.
Microorganisms ; 11(6)2023 Jun 10.
Article in English | MEDLINE | ID: mdl-37375055

ABSTRACT

Human milk oligosaccharides (HMOs) shape the developing infant gut microbiota. In this study, a semi-continuous colon simulator was used to evaluate the effect of 2 HMOs-2'-fucosyllactose (2'-FL) and 3-fucosyllactose (3-FL)-on the composition of infant faecal microbiota and microbial metabolites. The simulations were performed with and without a probiotic Bifidobacterium longum subspecies infantis Bi-26 (Bi-26) and compared with a control that lacked an additional carbon source. The treatments with HMOs decreased α-diversity and increased Bifidobacterium species versus the control, but the Bifidobacterium species differed between simulations. The levels of acetic acid and the sum of all short-chain fatty acids (SCFAs) trended toward an increase with 2'-FL, as did lactic acid with 2'-FL and 3-FL, compared with control. A clear correlation was seen between the consumption of HMOs and the increase in SCFAs (-0.72) and SCFAs + lactic acid (-0.77), whereas the correlation between HMO consumption and higher total bifidobacterial numbers was moderate (-0.46). Bi-26 decreased propionic acid levels with 2'-FL. In conclusion, whereas infant faecal microbiota varied between infant donors, the addition of 2'-FL and 3-FL, alone or in combination, increased the relative abundance and numbers Bifidobacterium species in the semi-continuous colon simulation model, correlating with the production of microbial metabolites. These findings may suggest that HMOs and probiotics benefit the developing infant gut microbiota.

6.
J Agric Food Chem ; 67(41): 11396-11402, 2019 Oct 16.
Article in English | MEDLINE | ID: mdl-31537068

ABSTRACT

Phytase is commonly used as a feed enzyme in monogastric animals to increase the bioavailability of phytate phosphorus and other nutrients. The accumulation of myo-inositol phosphate intermediates during phytate degradation in various segments of the gastrointestinal tract (GIT) is poorly understood. The aim of this study was to determine the efficacy of Buttiauxella spp. phytase in degrading the phytate in corn, soybean meal, and complete corn-soybean meal diet to myo-inositol phosphate esters (IP1-IP5) and completely dephosphorylated myo-inositol rings using an in vitro model of the poultry upper GIT. Our results show that the phytase hydrolyzes phytate efficiently to small IP esters, whereas the myo-inositol level remains constant between control and phytase treatments. Although the in vitro digestion model does not incorporate all factors that govern phytate hydrolysis, it is a valuable tool for evaluating phytase efficacy at various enzyme doses and with different feed ingredients.


Subject(s)
6-Phytase/chemistry , Animal Feed/analysis , Enterobacteriaceae/enzymology , Esters/chemistry , Inositol Phosphates/chemistry , Phytic Acid/chemistry , 6-Phytase/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Chickens , Digestion , Esters/metabolism , Food Additives/chemistry , Food Additives/metabolism , Gastrointestinal Tract/metabolism , Hydrolysis , Inositol Phosphates/metabolism , Models, Biological , Phytic Acid/metabolism , Glycine max/chemistry , Glycine max/metabolism , Zea mays/chemistry , Zea mays/metabolism
7.
Sci Rep ; 9(1): 13232, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31520068

ABSTRACT

Human milk oligosaccharides (HMOs) shape gut microbiota during infancy by acting as fermentable energy source. Using a semi-continuous colon simulator, effect of an HMO, 2'-fucosyllactose (2'-FL), on composition of the infant microbiota and microbial metabolites was evaluated in comparison to galacto-oligosaccharide (GOS) and lactose and control without additional carbon source. Data was analysed according to faecal sample donor feeding type: breast-fed (BF) or formula-fed (FF), and to rate of 2'-FL fermentation: fast or slow. Variation was found between the simulations in the ability to utilise 2'-FL. The predominant phyla regulated by 2'-FL, GOS and lactose were significant increase in Firmicutes, numerical in Actinobacteria, and numerical decrease in Proteobacteria compared to control. Verrucomicrobia increased in FF accounted for Akkermansia, whereas in fast-fermenting simulations Actinobacteria increased with trend for higher Bifidobacterium, and Proteobacteria decrease accounted for Enterobacteriaceae. Short-chain fatty acids and lactic acid with 2'-FL were produced in intermediate levels being between ones generated by the control and GOS or lactose. In 2'-FL fast-fermenting group, acetic acid specifically increased with 2'-FL, whereas lactose and GOS also increased lactic acid. The results highlight specificity of 2'-FL as energy source for only certain microbes over GOS and lactose in the simulated gut model.


Subject(s)
Colon/metabolism , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Lactose/pharmacology , Milk, Human/chemistry , Oligosaccharides/pharmacology , Trisaccharides/pharmacology , Colon/drug effects , Fermentation , Galactose/chemistry , Humans , Infant , Infant Formula/chemistry , Pilot Projects , Prebiotics/administration & dosage , Sweetening Agents/pharmacology
8.
J Proteome Res ; 17(3): 1041-1053, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29359944

ABSTRACT

The present study introduces a novel triple-phase (liquids, solids, and gases) approach, which employed uniformly labeled [U-13C] polydextrose (PDX) for the selective profiling of metabolites generated from dietary fiber fermentation in an in vitro colon simulator using human fecal inocula. Employing 13C NMR spectroscopy, [U-13C] PDX metabolism was observed from colonic digest samples. The major 13C-labeled metabolites generated were acetate, butyrate, propionate, and valerate. In addition to these short-chain fatty acids (SCFAs), 13C-labeled lactate, formate, succinate, and ethanol were detected in the colon simulator samples. Metabolite formation and PDX substrate degradation were examined comprehensively over time (24 and 48 h). Correlation analysis between 13C NMR spectra and gas production confirmed the anaerobic fermentation of PDX to SCFAs. In addition, 16S rRNA gene analysis showed that the level of Erysipelotrichaceae was influenced by PDX supplementation and Erysipelotrichaceae level was statistically correlated with SCFA formation. Overall, our study demonstrates a novel approach to link substrate fermentation and microbial function directly in a simulated colonic environment.


Subject(s)
Colon/metabolism , Fatty Acids, Volatile/metabolism , Feces/microbiology , Glucans/metabolism , Metabolome , Anaerobiosis , Bioreactors , Biotransformation , Carbon Isotopes , Colon/microbiology , Dietary Fiber/administration & dosage , Erysipelothrix/isolation & purification , Erysipelothrix/metabolism , Ethanol/metabolism , Fermentation , Formates/metabolism , Gastrointestinal Microbiome/physiology , Humans , Lactic Acid/metabolism , Magnetic Resonance Spectroscopy , Microbial Consortia/physiology , RNA, Ribosomal, 16S/genetics , Succinic Acid/metabolism
9.
EBioMedicine ; 13: 190-200, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27810310

ABSTRACT

BACKGROUND: The gut microbiota is interlinked with obesity, but direct evidence of effects of its modulation on body fat mass is still scarce. We investigated the possible effects of Bifidobacterium animalisssp. lactis 420 (B420) and the dietary fiber Litesse® Ultra polydextrose (LU) on body fat mass and other obesity-related parameters. METHODS: 225 healthy volunteers (healthy, BMI 28-34.9) were randomized into four groups (1:1:1:1), using a computer-generated sequence, for 6months of double-blind, parallel treatment: 1) Placebo, microcrystalline cellulose, 12g/d; 2) LU, 12g/d; 3) B420, 1010CFU/d in microcrystalline cellulose, 12g/d; 4) LU+B420, 12g+1010CFU/d. Body composition was monitored with dual-energy X-ray absorptiometry, and the primary outcome was relative change in body fat mass, comparing treatment groups to Placebo. Other outcomes included anthropometric measurements, food intake and blood and fecal biomarkers. The study was registered in Clinicaltrials.gov (NCT01978691). FINDINGS: There were marked differences in the results of the Intention-To-Treat (ITT; n=209) and Per Protocol (PP; n=134) study populations. The PP analysis included only those participants who completed the intervention with >80% product compliance and no antibiotic use. In addition, three participants were excluded from DXA analyses for PP due to a long delay between the end of intervention and the last DXA measurement. There were no significant differences between groups in body fat mass in the ITT population. However, LU+B420 and B420 seemed to improve weight management in the PP population. For relative change in body fat mass, LU+B420 showed a-4.5% (-1.4kg, P=0.02, N=37) difference to the Placebo group, whereas LU (+0.3%, P=1.00, N=35) and B420 (-3.0%, P=0.28, N=24) alone had no effect (overall ANOVA P=0.095, Placebo N=35). A post-hoc factorial analysis was significant for B420 (-4.0%, P=0.002 vs. Placebo). Changes in fat mass were most pronounced in the abdominal region, and were reflected by similar changes in waist circumference. B420 and LU+B420 also significantly reduced energy intake compared to Placebo. Changes in blood zonulin levels and hsCRP were associated with corresponding changes in trunk fat mass in the LU+B420 group and in the overall population. There were no differences between groups in the incidence of adverse events. DISCUSSION: This clinical trial demonstrates that a probiotic product with or without dietary fiber controls body fat mass. B420 and LU+B420 also reduced waist circumference and food intake, whereas LU alone had no effect on the measured outcomes.


Subject(s)
Cholera Toxin/blood , Dietary Fiber , Obesity/blood , Obesity/diet therapy , Overweight/blood , Overweight/diet therapy , Probiotics , Adipose Tissue/pathology , Adult , Biomarkers , Body Composition , Body Mass Index , Female , Gastrointestinal Microbiome , Haptoglobins , Healthy Volunteers , Humans , Male , Middle Aged , Obesity/pathology , Overweight/pathology , Protein Precursors , Treatment Outcome , Waist Circumference
10.
Front Nutr ; 3: 15, 2016.
Article in English | MEDLINE | ID: mdl-27376068

ABSTRACT

Obesity is related to the consumption of energy-dense foods in addition to changes in the microbiome where a higher abundance of gut Bacteroidetes can be found in lean subjects or after weight loss. Lactitol, a sweet-tasting sugar alcohol, is a common sugar-replacement in foods. Polydextrose (PDX), a highly branched glucose polymer, is known to reduce energy intake. Here, we test if the combined effects of lactitol or PDX in combination with Bacteroides species will have a beneficial metabolic response in rats fed a high-fat (HF) diet. A total of 175 male Wistar rats were fed either a LF or HF diet. Bacteroides thetaiotaomicron (10(10) bacteria/animal/day) was orally administered with or without lactitol (1.6-2 g/animal/day) or PDX (2 g/animal/day) for 8 days. Postprandial blood samples, cecal digesta, and feces were collected on the last day. Measurements included: body weight, feed consumption, cecal short-chain fatty acids, fecal dry matter and heat value, blood glucose, insulin, triglyceride, and satiety hormone concentrations. Lactitol and PDX decreased the mean body weight when administered with B. thetaiotaomicron or when lactitol was administered alone. Levels of postprandial plasma triglycerides declined with lactitol and PDX when administered with B. thetaiotaomicron. For intestinal hormone release, lactitol - alone or with B. thetaiotaomicron - increased the release of gastrointestinal peptide tyrosine tyrosine (PYY) as well as the area under the curve (AUC) measured for PYY (0-8 h). In addition, levels of insulin AUC (0-8 h) decreased in the lactitol and PDX-supplemented groups. Lactitol and PDX may both provide additional means to regulate postprandial metabolism and weight management, whereas the addition of B. thetaiotaomicron in the tested doses had only minor effects on the measured parameters.

11.
Food Chem Toxicol ; 92: 117-28, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27016492

ABSTRACT

AB-LIFE(®) is a probiotic product consisting of equal parts of three strains of Lactobacillus plantarum (CECT 7527, 7528, and 7529) blended with inert excipients. Whole genome sequencing was performed on each of the three strains. Antibiotic resistance was evaluated by genomic mining for resistance genes, and assessment for transferability. No risk of transfer potential was identified for any antibiotic resistance genes in the three strains. AB-LIFE(®) was evaluated for potential subchronic oral toxicity in rats, with dosages of 300 and 1000 mg/kg BW/day (equivalent to 5.55 × 10(10) and 1.85 × 10(11) CFU/kg BW/day). Survival of the three test strains through the gastrointestinal tract was supported by fecal analysis. No adverse effects were identified with respect to in-life parameters, clinical or anatomic pathology, translocation, or fecal chemical analyses. The no-observed-adverse-effect level (NOAEL) for AB-LIFE(®) in male and female rats was 1000 mg/kg BW/day (1.85 × 10(11) CFU of AB-LIFE(®)/kg BW/day), the highest dose level evaluated. These results, in conjunction with a previous acute toxicity study in rats, support the conclusion that AB-LIFE(®) is safe for human consumption.


Subject(s)
Drug Resistance, Microbial/genetics , Feces/microbiology , Gastrointestinal Tract/drug effects , Gene Expression Regulation, Bacterial/drug effects , Lactobacillus plantarum/physiology , Probiotics/toxicity , Toxicity Tests, Subchronic/methods , Administration, Oral , Animals , Feces/chemistry , Female , Genes, Bacterial/genetics , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Male , No-Observed-Adverse-Effect Level , Rats , Safety
12.
Biosci Biotechnol Biochem ; 76(6): 1135-9, 2012.
Article in English | MEDLINE | ID: mdl-22790935

ABSTRACT

Different ways of treating bran by baking enzymes prior to dough making and the baking process were used to increase the amount of water-soluble dietary fiber (DF) in wheat bread with added bran. Soluble DF was extracted from the bread with water and separated from the digestible material with gastrointestinal tract enzymes and by solvent precipitation. The baking enzyme mixtures tested (xylanase and glucanase/cellulase, with and without lipase) increased the amounts of soluble arabinoxylan and protein resistant to digestion. The isolated fiber was used as a growth substrate for 11 probiotic and intestinal Bifidobacterium strains, for commensal strains of Bacteroides fragilis and Escherichia coli, and for potential intestinal pathogenic strains of E. coli O157:H7, Salmonella typhimurium, and Clostridium perfringens. Fermentation analyses indicated that the tested strains had varying capacity to grow in the presence of the extracted fiber. Of the tested probiotic strains B. longum species generally showed the highest ability to utilize the fiber extracts, although the potential pathogens tested also showed an ability to grow on these fiber extracts. In sum, the enzymes used to improve the baking process for high-fiber bread can also be used to produce in situ soluble fiber material, which in turn can exert prebiotic effects on certain potentially beneficial microbes.


Subject(s)
Bread/microbiology , Dietary Fiber/metabolism , Probiotics/metabolism , Bacteroides fragilis/growth & development , Bacteroides fragilis/metabolism , Bifidobacterium/growth & development , Bifidobacterium/metabolism , Cellulases/metabolism , Clostridium perfringens/growth & development , Clostridium perfringens/metabolism , Endo-1,4-beta Xylanases/metabolism , Escherichia coli O157/growth & development , Escherichia coli O157/metabolism , Fermentation , Food Microbiology , Food Technology , Lipase/metabolism , Plant Proteins/biosynthesis , Xylans/biosynthesis
13.
Biosci Biotechnol Biochem ; 70(9): 2056-63, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16960357

ABSTRACT

A semi-continuous four-channel colon simulator was used to study the effects of lactose for the first time on the growth and fermentation dynamics of colonic microbiota. In six separate simulations, lactose supplementation increased the total SCFA concentration by 3-5 fold as compared with the baseline in the respective vessels. The total bacterial density was inversely correlated with lactic acid production (P = 0.003), while production of butyrate (P = 0.007) and propionate (P = 0.02) correlated with higher numbers of bacteria. A major shift in the microbial community structure in the lactose supplemented vessels was demonstrated by bacterial genomic %G+C-profiling of the total population, where lactose supplementation induced a clearly dominant peak in the bifidobacteria prominent area, %G+C 60-65. The transient shift to increased numbers of bifidobacteria (23-27%) of all bacteria in the first two vessels was also confirmed by the bifidobacteria-specific QPCR-method. In conclusion, lactose produced dramatic changes in microbiota composition and activity as compared with the baseline fermentation.


Subject(s)
Bifidobacterium/metabolism , Colon/microbiology , Lactose/pharmacology , Base Composition , Bifidobacterium/genetics , Biogenic Amines/metabolism , Bioreactors , Colon/drug effects , DNA, Bacterial/metabolism , Fatty Acids, Volatile/metabolism , Fermentation , Humans , Lactose/metabolism , Models, Anatomic
SELECTION OF CITATIONS
SEARCH DETAIL
...