Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Front Microbiol ; 14: 1273861, 2023.
Article in English | MEDLINE | ID: mdl-38075921

ABSTRACT

This study evaluated the effects of dietary supplementation with a postbiotic extract of Bifidobacterium breve BB091109 on pro-inflammatory cytokines levels and markers of endocrine function. A prospective, double-blind, placebo-controlled, randomized, single-centered, parallel study was conducted on a group of 40-55-year-old females. The study included 30 healthy females, divided into two groups: a supplement (n = 20) and a placebo (n = 10) groups. Blood and saliva samples were collected at baseline (wk0), after 4 weeks (wk 4) and 12 weeks (12wk) of daily supplementation (500 mg), and 4 weeks (wk 16) after termination of supplementation. The levels of fasting CRP, IL-6, IL-10, TNF-α, IFN-γ, DHEA, estradiol, estriol, progesterone, cortisol and human growth hormone were analysed. The results revealed a significant effect of the 90-day supplementation with B. breve postbiotic extract on changes in CRP, IL-6 levels, DHEA, estradiol and estriol. In conclusion, the supplementation with the B. breve postbiotic extract improved endocrine function in females over 40 years old and induced protective changes in inflammatory markers. These findings highlight the potential health benefits of this supplementation in promoting hormonal balance and reducing inflammation in this population.

2.
Cells ; 11(10)2022 05 18.
Article in English | MEDLINE | ID: mdl-35626706

ABSTRACT

The leading cause of gastroenteritis among young children worldwide is the Group A rotaviruses (RV), which produce a wide range of symptoms, from a limited diarrhea to severe dehydration and even death. After an RV infection, immunity is not complete and less severe re-infections usually occur. These infections could be ameliorated by nutritional interventions with bioactive compounds, such as prebiotics. The aim of this research was to study the impact of a particular galactooligosaccharide (B-GOS) on the RV symptomatology and immune response during two consecutive infections. Lewis neonatal rats were inoculated with SA11 (first RV infection) on day 6 of life and with EDIM (second RV infection) on day 17 of life. B-GOS group was administered by oral gavage with a daily dose of B-GOS between days three to nine of life. Clinical and immunological variables were assessed during both infective processes. In the first infection, after the prebiotic intervention with B-GOS, a lower incidence, duration, and overall severity of the diarrhea (p < 0.05) was observed. In addition, it improved another severity indicator, the fecal weight output, during the diarrhea period (p < 0.05). The second RV infection failed in provoking diarrhea in the groups studied. The immune response during first infection with SA11 was not affected by B-GOS administration and had no impact on second infection, but the prebiotic intervention significantly increased IFN-γ and TNF-α intestinal production after the second infection (p < 0.05). In summary, B-GOS supplementation is able to reduce the incidence and severity of the RV-associated diarrhea and to influence the immune response against RV infections.


Subject(s)
Rotavirus Infections , Rotavirus , Animals , Diarrhea/drug therapy , Intestines , Rats , Rats, Inbred Lew , Rotavirus/physiology , Rotavirus Infections/drug therapy
3.
Trends Microbiol ; 29(8): 667-685, 2021 08.
Article in English | MEDLINE | ID: mdl-33551269

ABSTRACT

Recent and ongoing developments in microbiome science are enabling new frontiers of research for probiotics and prebiotics. Novel types, mechanisms, and applications currently under study have the potential to change scientific understanding as well as nutritional and healthcare applications of these interventions. The expansion of related fields of microbiome-targeted interventions, and an evolving landscape for implementation across regulatory, policy, prescriber, and consumer spheres, portends an era of significant change. In this review we examine recent, emerging, and anticipated trends in probiotic and prebiotic science, and create a vision for broad areas of developing influence in the field.


Subject(s)
Bacteria/genetics , Gastrointestinal Microbiome/physiology , Prebiotics , Probiotics , Bacteria/metabolism , Gastrointestinal Microbiome/genetics , Humans , Precision Medicine/methods , Precision Medicine/trends
4.
Microbiome ; 6(1): 133, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30071894

ABSTRACT

BACKGROUND: Different dietary approaches, such as gluten and casein free diets, or the use of probiotics and prebiotics have been suggested in autistic spectrum disorders in order to reduce gastrointestinal (GI) disturbances. GI symptoms are of particular interest in this population due to prevalence and correlation with the severity of behavioural traits. Nowadays, there is lack of strong evidence about the effect of dietary interventions on these problems, particularly prebiotics. Therefore, we assessed the impact of exclusion diets and a 6-week Bimuno® galactooligosaccharide (B-GOS®) prebiotic intervention in 30 autistic children. RESULTS: The results showed that children on exclusion diets reported significantly lower scores of abdominal pain and bowel movement, as well as lower abundance of Bifidobacterium spp. and Veillonellaceae family, but higher presence of Faecalibacterium prausnitzii and Bacteroides spp. In addition, significant correlations were found between bacterial populations and faecal amino acids in this group, compared to children following an unrestricted diet. Following B-GOS® intervention, we observed improvements in anti-social behaviour, significant increase of Lachnospiraceae family, and significant changes in faecal and urine metabolites. CONCLUSIONS: To our knowledge, this is the first study where the effect of exclusion diets and prebiotics has been evaluated in autism, showing potential beneficial effects. A combined dietary approach resulted in significant changes in gut microbiota composition and metabolism suggesting that multiple interventions might be more relevant for the improvement of these aspects as well as psychological traits. TRIAL REGISTRATION: NCT02720900 ; registered in November 2015.


Subject(s)
Autism Spectrum Disorder/diet therapy , Bacteria/classification , Gastrointestinal Tract/microbiology , Prebiotics/administration & dosage , Autism Spectrum Disorder/microbiology , Autism Spectrum Disorder/urine , Bacteria/isolation & purification , Child , Child, Preschool , Double-Blind Method , Feces/chemistry , Feces/microbiology , Female , Humans , Male , Urine/chemistry , Urine/microbiology
5.
Neurogastroenterol Motil ; 30(11): e13440, 2018 11.
Article in English | MEDLINE | ID: mdl-30109908

ABSTRACT

BACKGROUND: Prebiotics exert beneficial effects upon gastrointestinal (GI) environment, but this is not always accompanied with a positive effect on GI symptoms. B-GOS® is a prebiotic with high selectivity toward bifidobacteria and a variety of other beneficial effects in humans. Here, we investigated its effect on GI symptoms in adults who suffer with bloating, abdominal pain, and flatulence. METHODS: In a double-blind, placebo-controlled, crossover study, 83 subjects from the general population who presented with GI symptoms during screening period and had a predicted probability of functional bowel disorder of more than 75% were randomized to receive either a placebo or the B-GOS® treatment (2.75 g/d). Subjects were screened for the presence of GI symptoms for 1 week, they consumed the treatments for 2 weeks, and then went through a 2-week washout period, before switching to the other treatment for the final 2 weeks. GI symptoms, bowel movements, and stool consistency were assessed in daily and weekly questionnaires. Quality of life was assessed weekly and depression and anxiety at the end of each treatment period. RESULTS: B-GOS® resulted in significantly (P < 0.001) lower scores for bloating, flatulence, and abdominal pain both from baseline and placebo at the end of first week. The effect was sustained at the end of second week. It had no effect on the number of bowel movements, consistency of stools, quality of life, or mood throughout the study. CONCLUSION: Results suggest that B-GOS® could possibly be used in the management of bloating, flatulence, or abdominal pain and warrant further investigation.


Subject(s)
Abdominal Pain , Flatulence , Prebiotics , Adult , Cross-Over Studies , Dietary Supplements , Double-Blind Method , Female , Galactose/pharmacology , Humans , Male
6.
Gastroenterology ; 155(4): 1004-1007, 2018 10.
Article in English | MEDLINE | ID: mdl-29964041

ABSTRACT

Prebiotics and diets low in fermentable oligo-, di-, mono-saccharides and polyols (low-FODMAP diet) might reduce symptoms in patients with functional gastrointestinal disorders, despite reports that some nonabsorbable, fermentable meal products (prebiotics) provide substrates for colonic bacteria and thereby increase gas production. We performed a randomized, parallel, double-blind study of patients with functional gastrointestinal disorders with flatulence. We compared the effects of a prebiotic supplement (2.8 g/d Bimuno containing 1.37 g beta-galactooligosaccharide) plus a placebo (Mediterranean-type diet (prebiotic group, n = 19) vs a placebo supplement (2.8 g xylose) plus a diet low in FODMAP (low-FODMAP group, n = 21) for 4 weeks; patients were then followed for 2 weeks. The primary outcome was effects on composition of the fecal microbiota, analyzed by 16S sequencing. Secondary outcomes were intestinal gas production and digestive sensations. After 4 weeks, we observed opposite effects on microbiota in each group, particularly in relation to the abundance of Bifidobacterium sequences (increase in the prebiotic group and decrease in the low-FODMAP group; P = .042), and Bilophila wadsworthia (decrease in the prebiotic group and increase in the low-FODMAP group; P = .050). After 4 weeks, both groups had statistically significant reductions in all symptom scores, except reductions in flatulence and borborygmi were not significant in the prebiotic group. Although the decrease in symptoms persisted for 2 weeks after patients discontinued prebiotic supplementation, symptoms reappeared immediately after patients discontinued the low-FODMAP diet. Intermittent prebiotic administration might therefore be an alternative to dietary restrictions for patients with functional gut symptoms. ClinicalTrials.gov no.: NCT02210572.


Subject(s)
Bacteria/metabolism , Diet, Carbohydrate-Restricted , Dietary Carbohydrates/administration & dosage , Fermentation , Gastrointestinal Diseases/diet therapy , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Prebiotics , Diet, Carbohydrate-Restricted/adverse effects , Dietary Carbohydrates/adverse effects , Dietary Carbohydrates/metabolism , Double-Blind Method , Europe , Gastrointestinal Diseases/diagnosis , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/microbiology , Humans , Prebiotics/adverse effects , Recurrence , Remission Induction , Time Factors , Treatment Outcome
7.
FEMS Microbiol Ecol ; 93(2)2017 02.
Article in English | MEDLINE | ID: mdl-27856622

ABSTRACT

Children with autism spectrum disorders (ASD) often suffer gastrointestinal problems consistent with imbalances in the gut microbial population. Treatment with antibiotics or pro/prebiotics has been postulated to regulate microbiota and improve gut symptoms, but there is a lack of evidence for such approaches, especially for prebiotics. This study assessed the influence of a prebiotic galactooligosaccharide (B-GOS) on gut microbial ecology and metabolic function using faecal samples from autistic and non-autistic children in an in vitro gut model system. Bacteriology was analysed using flow cytometry combined with fluorescence in situ hybridization and metabolic activity by HPLC and 1H-NMR. Consistent with previous studies, the microbiota of children with ASD contained a higher number of Clostridium spp. and a lower number of bifidobacteria compared with non-autistic children. B-GOS administration significantly increased bifidobacterial populations in each compartment of the models, both with autistic and non-autistic-derived samples, and lactobacilli in the final vessel of non-autistic models. In addition, changes in other bacterial population have been seen in particular for Clostridium, Rosburia, Bacteroides, Atopobium, Faecalibacterium prausnitzii, Sutterella spp. and Veillonellaceae. Furthermore, the addition of B-GOS to the models significantly altered short-chain fatty acid production in both groups, and increased ethanol and lactate in autistic children.


Subject(s)
Autistic Disorder/microbiology , Child , Feces/microbiology , Fermentation , Prebiotics , Anti-Bacterial Agents , Bacteroides/metabolism , Bifidobacterium/growth & development , Bifidobacterium/metabolism , Clostridium/metabolism , Fatty Acids, Volatile/metabolism , Humans , In Situ Hybridization, Fluorescence , Lactic Acid , Lactobacillus/metabolism , Microbiota , Oligosaccharides/metabolism
8.
Br J Nutr ; 116(5): 798-804, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27523186

ABSTRACT

Gut microbes have a substantial influence on systemic immune function and allergic sensitisation. Manipulation of the gut microbiome through prebiotics may provide a potential strategy to influence the immunopathology of asthma. This study investigated the effects of prebiotic Bimuno-galactooligosaccharide (B-GOS) supplementation on hyperpnoea-induced bronchoconstriction (HIB), a surrogate for exercise-induced bronchoconstriction, and airway inflammation. A total of ten adults with asthma and HIB and eight controls without asthma were randomised to receive 5·5 g/d of either B-GOS or placebo for 3 weeks separated by a 2-week washout period. The peak fall in forced expiratory volume in 1 s (FEV1) following eucapnic voluntary hyperpnoea (EVH) defined HIB severity. Markers of airway inflammation were measured at baseline and after EVH. Pulmonary function remained unchanged in the control group. In the HIB group, the peak post-EVH fall in FEV1 at day 0 (-880 (sd 480) ml) was unchanged after placebo, but was attenuated by 40 % (-940 (sd 460) v. -570 (sd 310) ml, P=0·004) after B-GOS. In the HIB group, B-GOS reduced baseline chemokine CC ligand 17 (399 (sd 140) v. 323 (sd 144) pg/ml, P=0·005) and TNF-α (2·68 (sd 0·98) v. 2·18 (sd 0·59) pg/ml, P=0·040) and abolished the EVH-induced 29 % increase in TNF-α. Baseline C-reactive protein was reduced following B-GOS in HIB (2·46 (sd 1·14) v. 1·44 (sd 0·41) mg/l, P=0·015) and control (2·16 (sd 1·02) v. 1·47 (sd 0·33) mg/l, P=0·050) groups. Chemokine CC ligand 11 and fraction of exhaled nitric oxide remained unchanged. B-GOS supplementation attenuated airway hyper-responsiveness with concomitant reductions in markers of airway inflammation associated with HIB.


Subject(s)
Bronchoconstriction , Inflammation/prevention & control , Oligosaccharides/pharmacology , Prebiotics/administration & dosage , Respiratory Tract Diseases/prevention & control , Adult , Biomarkers , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , Case-Control Studies , Chemokines/blood , Chemokines/genetics , Chemokines/metabolism , Cross-Over Studies , Double-Blind Method , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Immunoglobulin E/blood , Immunoglobulin E/genetics , Immunoglobulin E/metabolism , Inflammation/etiology , Male , Oligosaccharides/administration & dosage , Respiratory Tract Diseases/etiology , Tumor Necrosis Factor-alpha/blood , Young Adult
9.
Br J Nutr ; 116(3): 480-6, 2016 08.
Article in English | MEDLINE | ID: mdl-27267934

ABSTRACT

Prebiotic oligosaccharides have the ability to generate important changes in the gut microbiota composition that may confer health benefits to the host. Reducing the impurities in prebiotic mixtures could expand their applications in food industries and improve their selectivity and prebiotic effect on the potential beneficial bacteria such as bifidobacteria and lactobacilli. This study aimed to determine the in vitro potential fermentation properties of a 65 % galacto-oligosaccharide (GOS) content Bimuno® GOS (B-GOS) on gut microbiota composition and their metabolites. Fermentation of 65 % B-GOS was compared with 52 % B-GOS in pH- and volume-controlled dose-response anaerobic batch culture experiments. In total, three different doses (1, 0·5 and 0·33 g equivalent to 0·1, 0·05 and 0·033 g/l) were tested. Changes in the gut microbiota during a time course were identified by fluorescence in situ hybridisation, whereas small molecular weight metabolomics profiles and SCFA were determined by 1H-NMR analysis and GC, respectively. The 65 % B-GOS showed positive modulation of the microbiota composition during the first 8 h of fermentation with all doses. Administration of the specific doses of B-GOS induced a significant increase in acetate as the major SCFA synthesised compared with propionate and butyrate concentrations, but there were no significant differences between substrates. The 65 % B-GOS in syrup format seems to have, in all the analysis, an efficient prebiotic effect. However, the applicability of such changes remains to be shown in an in vivo trial.


Subject(s)
Bacteria/drug effects , Fatty Acids, Volatile/metabolism , Fermentation , Galactose/pharmacology , Gastrointestinal Microbiome/drug effects , Oligosaccharides/pharmacology , Prebiotics , Acetic Acid/metabolism , Bacteria/growth & development , Bacteria/metabolism , Bifidobacterium/drug effects , Bifidobacterium/growth & development , Bifidobacterium/metabolism , Butyric Acid/metabolism , Colon/metabolism , Colon/microbiology , Feces , Humans , Lactobacillus/drug effects , Lactobacillus/growth & development , Lactobacillus/metabolism , Metabolomics , Propionates/metabolism
10.
Br J Nutr ; 114(4): 586-95, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26218845

ABSTRACT

It is recognised that ageing induces various changes to the human colonic microbiota. Most relevant is a reduction in bifidobacteria, which is a health-positive genus. Prebiotics, such as galacto-oligosaccharides (GOS), are dietary ingredients that selectively fortify beneficial gut microbial groups. Therefore, they have the potential to reverse the age-related decline in bifidobacteria and modulate associated health parameters. We assessed the effect of GOS mixture (Bimuno (B-GOS)) on gut microbiota, markers of immune function and metabolites in forty elderly (age 65-80 years) volunteers in a randomised, double-blind, placebo (maltodextrin)-controlled, cross-over study. The intervention periods consisted of 10 weeks with daily doses of 5·5 g/d with a 4-week washout period in between. Blood and faecal samples were collected for the analyses of faecal bacterial populations and immune and metabolic biomarkers. B-GOS consumption led to significant increases in bacteroides and bifidobacteria, the latter correlating with increased lactic acid in faecal waters. Higher IL-10, IL-8, natural killer cell activity and C-reactive protein and lower IL-1ß were also observed. Administration of B-GOS to elderly volunteers may be useful in positively affecting the microbiota and some markers of immune function associated with ageing.


Subject(s)
Aging/immunology , Bacteria/growth & development , Gastrointestinal Tract/drug effects , Immune System/drug effects , Microbiota/drug effects , Oligosaccharides/pharmacology , Prebiotics , Aged , Aged, 80 and over , Aging/metabolism , Bacteroides/growth & development , Bifidobacterium/growth & development , Biomarkers/blood , C-Reactive Protein/metabolism , Cross-Over Studies , Double-Blind Method , Feces/microbiology , Female , Galactose/pharmacology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Humans , Immune System/metabolism , Immune System/microbiology , Interleukins/blood , Killer Cells, Natural/metabolism , Lactic Acid/metabolism , Male , Metabolomics
11.
Gut Microbes ; 6(5): 300-9, 2015.
Article in English | MEDLINE | ID: mdl-26099034

ABSTRACT

Prebiotics are dietary carbohydrates that favourably modulate the gut microbiota. The aims of the present study were to develop a functional prebiotic bread using Bimuno®, (galactooligosaccharide (B-GOS) mixture), for modulation of the gut microbiota in vitro in individuals at risk of metabolic syndrome. A control bread, (no added prebiotic) and positive control bread (containing equivalent carbohydrate to B-GOS bread) were also developed. A 3-stage continuous in vitro colonic model was used to assess prebiotic functionality of the breads. Bacteria were quantified by fluorescence in situ hybridization and short chain fatty acids by gas chromatography. Ion-exchange chromatography was used to determine GOS concentration after bread production. Following B-GOS bread fermentation numbers of bifidobacteria and lactobacilli were significantly higher compared to controls. There was no significant degradation of B-GOS during bread manufacture, indicating GOS withstood the manufacturing process. Furthermore, based on previous research, increased bifidobacteria and butyrate levels could be of benefit to those with obesity related conditions. Our findings support utilization of prebiotic enriched bread for improving gastrointestinal health.


Subject(s)
Bread , Colon/chemistry , Colon/microbiology , Diet Therapy/methods , Drug Carriers/administration & dosage , Metabolic Syndrome/therapy , Prebiotics/administration & dosage , Bacterial Load , Bifidobacterium/isolation & purification , Chromatography, Gas , Chromatography, Ion Exchange , Fatty Acids, Volatile/analysis , In Situ Hybridization, Fluorescence , Lactobacillus/isolation & purification , Models, Biological , Oligosaccharides/analysis
12.
PLoS One ; 10(3): e0121955, 2015.
Article in English | MEDLINE | ID: mdl-25807417

ABSTRACT

A three-stage continuous fermentative colonic model system was used to monitor in vitro the effect of different orange juice formulations on prebiotic activity. Three different juices with and without Bimuno, a GOS mixture containing galactooligosaccharides (B-GOS) were assessed in terms of their ability to induce a bifidogenic microbiota. The recipe development was based on incorporating 2.75g B-GOS into a 250 ml serving of juice (65°Brix of concentrate juice). Alongside the production of B-GOS juice, a control juice--orange juice without any additional Bimuno and a positive control juice, containing all the components of Bimuno (glucose, galactose and lactose) in the same relative proportions with the exception of B-GOS were developed. Ion Exchange Chromotography analysis was used to test the maintenance of bimuno components after the production process. Data showed that sterilisation had no significant effect on concentration of B-GOS and simple sugars. The three juice formulations were digested under conditions resembling the gastric and small intestinal environments. Main bacterial groups of the faecal microbiota were evaluated throughout the colonic model study using 16S rRNA-based fluorescence in situ hybridization (FISH). Potential effects of supplementation of the juices on microbial metabolism were studied measuring short chain fatty acids (SCFAs) using gas chromatography. Furthermore, B-GOS juices showed positive modulations of the microbiota composition and metabolic activity. In particular, numbers of faecal bifidobacteria and lactobacilli were significantly higher when B-GOS juice was fermented compared to controls. Furthermore, fermentation of B-GOS juice resulted in an increase in Roseburia subcluster and concomitantly increased butyrate production, which is of potential benefit to the host. In conclusion, this study has shown B-GOS within orange juice can have a beneficial effect on the fecal microbiota.


Subject(s)
Bifidobacterium , Citrus sinensis , Colon/microbiology , Microbiota/drug effects , Models, Biological , Prebiotics/analysis , Adult , Female , Fermentation , Humans , Male
13.
Br J Nutr ; 111(5): 773-84, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24128654

ABSTRACT

The gut microbiota plays an important role in the development of the immune and gastrointestinal systems of infants. In the present study, we investigated whether increased salmon consumption during pregnancy, maternal weight gain during pregnancy or mode of infant feeding alter the markers of gut immune defence and inflammation. Women (n 123) who rarely ate oily fish were randomly assigned to continue consuming their habitual diet or to consume two 150 g portions of farmed salmon per week from 20 weeks of pregnancy to delivery. Faecal samples were collected from the mothers (n 75) at 38 weeks of gestation and from their infants (n 38) on days 7, 14, 28 and 84 post-partum. Fluorescence in situ hybridisation was used to determine faecal microbiota composition and ELISA to measure faecal secretory IgA (sIgA) and calprotectin concentrations. There was no effect of salmon consumption on maternal faecal microbiota or on maternal or infant faecal sIgA and calprotectin concentrations. The degree of weight gain influenced maternal faecal microbiota, and the mode of infant feeding influenced infant faecal microbiota. Faecal samples collected from infants in the salmon group tended to have lower bacterial counts of the Atopobium cluster compared with those collected from infants in the control group (P=0·097). This difference was significant in the formula-fed infants (P< 0·05), but not in the exclusively breast-fed infants. In conclusion, the impact of oily fish consumption during pregnancy on maternal and infant gut microbiota composition is limited, but significant differences are associated with maternal weight gain during pregnancy and mode of infant feeding.


Subject(s)
Immunity, Mucosal , Immunoglobulin A, Secretory/analysis , Intestines/microbiology , Leukocyte L1 Antigen Complex/analysis , Prenatal Nutritional Physiological Phenomena , Salmon , Seafood , Adult , Animals , Biomarkers/analysis , Child Development , Feces/chemistry , Feces/microbiology , Female , Humans , Hypersensitivity/epidemiology , Hypersensitivity/prevention & control , Infant Nutritional Physiological Phenomena , Infant, Newborn , Intestines/growth & development , Intestines/immunology , Pregnancy , Risk , Seafood/adverse effects , Single-Blind Method , United Kingdom/epidemiology , Weight Gain
14.
J Nutr ; 143(3): 324-31, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23303873

ABSTRACT

Metabolic syndrome is a set of disorders that increases the risk of developing cardiovascular disease. The gut microbiota is altered toward a less beneficial composition in overweight adults and this change can be accompanied by inflammation. Prebiotics such as galactooligosaccharides can positively modify the gut microbiota and immune system; some may also reduce blood lipids. We assessed the effect of a galactooligosaccharide mixture [Bi2muno (B-GOS)] on markers of metabolic syndrome, gut microbiota, and immune function in 45 overweight adults with ≥3 risk factors associated with metabolic syndrome in a double-blind, randomized, placebo (maltodextrin)-controlled, crossover study (with a 4-wk wash-out period between interventions). Whole blood, saliva, feces, and anthropometric measurements were taken at the beginning, wk 6, and end of each 12-wk intervention period. Predominant groups of fecal bacteria were quantified and full blood count, markers of inflammation and lipid metabolism, insulin, and glucose were measured. B-GOS increased the number of fecal bifidobacteria at the expense of less desirable groups of bacteria. Increases in fecal secretory IgA and decreases in fecal calprotectin, plasma C-reactive protein, insulin, total cholesterol (TC), TG, and the TC:HDL cholesterol ratio were also observed. Administration of B-GOS to overweight adults resulted in positive effects on the composition of the gut microbiota, the immune response, and insulin, TC, and TG concentrations. B-GOS may be a useful candidate for the enhancement of gastrointestinal health, immune function, and the reduction of metabolic syndrome risk factors in overweight adults.


Subject(s)
Bacteria/drug effects , Galactose/therapeutic use , Immunity/drug effects , Metabolic Syndrome/drug therapy , Oligosaccharides/therapeutic use , Overweight/drug therapy , Prebiotics , Adult , Bacteria/growth & development , Bifidobacterium/drug effects , Bifidobacterium/growth & development , Biomarkers/metabolism , C-Reactive Protein/metabolism , Cross-Over Studies , Double-Blind Method , Feces/chemistry , Feces/microbiology , Female , Galactose/pharmacology , Humans , Immunoglobulin A/metabolism , Inflammation/blood , Inflammation/prevention & control , Insulin/blood , Leukocyte L1 Antigen Complex/metabolism , Lipids/blood , Male , Metabolic Syndrome/metabolism , Metagenome/drug effects , Middle Aged , Oligosaccharides/pharmacology , Overweight/immunology , Overweight/metabolism , Overweight/microbiology , Risk Factors
15.
Am J Clin Nutr ; 93(1): 62-72, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21068351

ABSTRACT

BACKGROUND: The absorption of cocoa flavanols in the small intestine is limited, and the majority of the flavanols reach the large intestine where they may be metabolized by resident microbiota. OBJECTIVE: We assessed the prebiotic potential of cocoa flavanols in a randomized, double-blind, crossover, controlled intervention study. DESIGN: Twenty-two healthy human volunteers were randomly assigned to either a high-cocoa flavanol (HCF) group (494 mg cocoa flavanols/d) or a low-cocoa flavanol (LCF) group (23 mg cocoa flavanols/d) for 4 wk. This was followed by a 4-wk washout period before volunteers crossed to the alternant arm. Fecal samples were recovered before and after each intervention, and bacterial numbers were measured by fluorescence in situ hybridization. A number of other biochemical and physiologic markers were measured. RESULTS: Compared with the consumption of the LCF drink, the daily consumption of the HCF drink for 4 wk significantly increased the bifidobacterial (P < 0.01) and lactobacilli (P < 0.001) populations but significantly decreased clostridia counts (P < 0.001). These microbial changes were paralleled by significant reductions in plasma triacylglycerol (P < 0.05) and C-reactive protein (P < 0.05) concentrations. Furthermore, changes in C-reactive protein concentrations were linked to changes in lactobacilli counts (P < 0.05, R(2) = -0.33 for the model). These in vivo changes were closely paralleled by cocoa flavanol-induced bacterial changes in mixed-batch culture experiments. CONCLUSION: This study shows, for the first time to our knowledge, that consumption of cocoa flavanols can significantly affect the growth of select gut microflora in humans, which suggests the potential prebiotic benefits associated with the dietary inclusion of flavanol-rich foods. This trial was registered at clinicaltrials.gov as NCT01091922.


Subject(s)
Bifidobacterium/drug effects , Cacao/chemistry , Flavonoids/pharmacology , Lactobacillus/drug effects , Prebiotics , Adult , Antioxidants/metabolism , Bifidobacterium/growth & development , Biomarkers , Blood Pressure , C-Reactive Protein/analysis , Cross-Over Studies , Diet , Double-Blind Method , Feces/microbiology , Female , Flavonoids/metabolism , Humans , Lactobacillus/growth & development , Male
16.
Am J Clin Nutr ; 88(5): 1438-46, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18996881

ABSTRACT

BACKGROUND: Aging is associated with reduced numbers of beneficial colonic bifidobacteria and impaired immunity. Galactooligosaccharides (GOSs) stimulate the growth of bifidobacteria in younger adults, but little is known about their effects in the elderly and their immunomodulatory capacity. OBJECTIVE: We assessed the effect of a prebiotic GOS mixture (B-GOS) on immune function and fecal microflora composition in healthy elderly subjects. DESIGN: In a double-blind, placebo-controlled, crossover study, 44 elderly subjects were randomly assigned to receive either a placebo or the B-GOS treatment (5.5 g/d). Subjects consumed the treatments for 10 wk, and then went through a 4-wk washout period, before switching to the other treatment for the final 10 wk. Blood and fecal samples were collected at the beginning, middle (5 wk), and end of the test period. Predominant bacterial groups were quantified, and phagocytosis, natural killer (NK) cell activity, cytokine production, plasma cholesterol, and HDL cholesterol were measured. RESULTS: B-GOS significantly increased the numbers of beneficial bacteria, especially bifidobacteria, at the expense of less beneficial groups compared with the baseline and placebo. Significant increases in phagocytosis, NK cell activity, and the production of antiinflammatory cytokine interleukin-10 (IL-10) and significant reduction in the production of proinflammatory cytokines (IL-6, IL-1beta, and tumor necrosis factor-alpha) were also observed. B-GOS exerted no effects on total cholesterol or HDL-cholesterol production, however. CONCLUSIONS: B-GOS administration to healthy elderly persons resulted in positive effects on both the microflora composition and the immune response. Therefore, B-GOS may be a useful dietary candidate for the enhancement of gastrointestinal health and immune function in elderly persons.


Subject(s)
Aging/immunology , Bifidobacterium/growth & development , Feces/microbiology , Oligosaccharides/administration & dosage , Oligosaccharides/metabolism , Probiotics , Aged , Aging/physiology , Analysis of Variance , Bifidobacterium/drug effects , Cholesterol/blood , Cholesterol, HDL/blood , Colony Count, Microbial , Cross-Over Studies , Cytokines/biosynthesis , Cytokines/immunology , Dietary Supplements , Double-Blind Method , Female , Humans , Killer Cells, Natural/immunology , Male , Middle Aged , Phagocytosis/immunology
17.
Adv Exp Med Biol ; 635: 79-92, 2008.
Article in English | MEDLINE | ID: mdl-18841705

ABSTRACT

Functional foods (specific nutrient and/or food components) should beneficially affect one or more target functions in the body. The use of functional foods as a form of preventive medicine has been the subject of much research over the last two decades. It is well known that nutrition plays a vital role in chronic diseases, but it is only recently that data relating to the effects of specific nutrients or foods on the immune system have become available. This chapter aims to summarize the effects of some functional foods (e.g., prebiotics and micronutrients) on the immune system. It should be noted, however, that studies into the role of functional foods with regard to the human immune system are still in their infancy and a great deal of controversy surrounds the health claims attributed to some functional foods. Consequently, thorough studies are required in human and animal systems if we are to move towards developing a functional diet that provides maximal health benefits.


Subject(s)
Diet , Food , Immunity , Colon/physiology , Dietary Fiber , Humans
18.
Am J Clin Nutr ; 87(3): 785-91, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18326619

ABSTRACT

BACKGROUND: Galactooligosaccharides are selectively fermented by the beneficial member of the colonic microflora contributing to the health of the host. OBJECTIVE: We assessed the prebiotic potential of a novel galactooligosaccharide produced through the action of beta-galactosidases, originating from a probiotic Bifidobacterium bifidum strain, against a galactooligosaccharide produced through the action of an industrial beta-galactosidase and a placebo. DESIGN: Fifty-nine healthy human volunteers participated in this study. Initially, the effect of the matrix on the prebiotic properties of a commercially available galactooligosaccharide (7 g/d) was assessed during 7-d treatment periods with a 7-d washout period in between. During the second phase, 30 volunteers were assigned to a sequence of treatments (7 d) differing in the amount of the novel galactooligosaccharide (0, 3.6, or 7 g/d). Stools were recovered before and after each intervention, and bacteria numbers were determined by fluorescent in situ hybridization. RESULTS: Addition of the novel galactooligosaccharide mixture significantly increased the bifidobacterial population ratio compared with the placebo (P < 0.05), whereas 7 g/d of the novel galactooligosaccharide significantly increased the bifidobacterial ratio compared with the commercial galactooligosaccharide (P < 0.05). Moreover, a significant relation (P < 0.001) between the bifidobacteria proportion and the novel galactooligosaccharide dose (0, 3.6, and 7 g/d) was observed. This relation was similar to the effect of the novel galactooligosaccharide on the prebiotic index of each dose. CONCLUSIONS: This study showed that galactooligosaccharide mixtures produced with different beta-galactosidases show different prebiotic properties and that, by using enzymes originating from bifidobacterial species, an increase in the bifidogenic properties of the prebiotic product is achievable.


Subject(s)
Bifidobacterium/enzymology , Galactose/metabolism , Oligosaccharides/metabolism , Probiotics , beta-Galactosidase/metabolism , Adult , Colon/metabolism , Colon/microbiology , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Feces/microbiology , Female , Fermentation , Galactose/biosynthesis , Humans , In Situ Hybridization, Fluorescence/methods , Male , Oligosaccharides/biosynthesis
19.
Br J Nutr ; 99(4): 782-92, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17977475

ABSTRACT

We have investigated the bacterial-dependent metabolism of ( - )-epicatechin and (+)-catechin using a pH-controlled, stirred, batch-culture fermentation system reflective of the distal region of the human large intestine. Incubation of ( - )-epicatechin or (+)-catechin (150 mg/l or 1000 mg/l) with faecal bacteria, led to the generation of 5-(3',4'-dihydroxyphenyl)-gamma-valerolactone, 5-phenyl-gamma-valerolactone and phenylpropionic acid. However, the formation of these metabolites from (+)-catechin required its initial conversion to (+)-epicatechin. The metabolism of both flavanols occurred in the presence of favourable carbon sources, notably sucrose and the prebiotic fructo-oligosaccharides, indicating that bacterial utilisation of flavanols also occurs when preferential energy sources are available. (+)-Catechin incubation affected the growth of select microflora, resulting in a statistically significant increase in the growth of the Clostridium coccoides-Eubacterium rectale group, Bifidobacterium spp. and Escherichia coli, as well as a significant inhibitory effect on the growth of the C. histolyticum group. In contrast, the effect of ( - )-epicatechin was less profound, only significantly increasing the growth of the C. coccoides-Eubacterium rectale group. These potential prebiotic effects for both (+)-catechin and ( - )-epicatechin were most notable at the lower concentration of 150 mg/l. As both ( - )-epicatechin and (+)-catechin were converted to the same metabolites, the more dramatic change in the growth of distinct microfloral populations produced by (+)-catechin incubation may be linked to the bacterial conversion of (+)-catechin to (+)-epicatechin. Together these data suggest that the consumption of flavanol-rich foods may support gut health through their ability to exert prebiotic actions.


Subject(s)
Antioxidants/pharmacology , Bacteria/metabolism , Feces/microbiology , Flavonoids/pharmacology , Antioxidants/metabolism , Bacteria/drug effects , Bacteriological Techniques , Bifidobacterium/growth & development , Catechin/metabolism , Catechin/pharmacology , Clostridium/growth & development , Escherichia coli/growth & development , Eubacterium/growth & development , Fermentation , Flavonoids/metabolism , Humans , Intestine, Large/metabolism , Intestine, Large/microbiology
20.
Anaerobe ; 13(5-6): 200-7, 2007.
Article in English | MEDLINE | ID: mdl-17611130

ABSTRACT

Prebiotics and probiotics are increasingly being used to produce potentially synbiotic foods, particularly through dairy products as vehicles. It is well known that both ingredients may offer benefits to improve the host health. This research aimed to evaluate the prebiotic potential of novel petit-suisse cheeses using an in vitro fermentation model. Five petit-suisse cheese formulations combining candidate prebiotics (inulin, oligofructose, honey) and probiotics (Lactobacillus acidophilus, Bifidobacterium lactis) were tested in vitro using sterile, stirred, batch culture fermentations with human faecal slurry. Measurement of prebiotic effect (MPE) values were generated comparing bacterial changes through determination of maximum growth rates of groups, rate of substrate assimilation and production of lactate and short chain fatty acids. Fastest fermentation and high lactic acid production, promoting increased growth rates of bifidobacteria and lactobacilli, were achieved with addition of prebiotics to a probiotic cheese (made using starter+probiotics). Addition of probiotic strains to control cheese (made using just a starter culture) also resulted in high lactic acid production. Highest MPE values were obtained with addition of prebiotics to a probiotic cheese, followed by addition of prebiotics and/or probiotics to a control cheese. Under the in vitro conditions used, cheese made with the combination of different prebiotics and probiotics resulted in the most promising functional petit-suisse cheese. The study allowed comparison of potentially functional petit-suisse cheeses and screening of preferred synbiotic potential for future market use.


Subject(s)
Bifidobacterium/growth & development , Cheese/microbiology , Food Microbiology , Lactobacillus/metabolism , Probiotics/metabolism , Cheese/analysis , Fermentation , Lactobacillus/growth & development , Research Design
SELECTION OF CITATIONS
SEARCH DETAIL
...