Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Nutrients ; 14(12)2022 Jun 19.
Article in English | MEDLINE | ID: mdl-35745275

ABSTRACT

BACKGROUND: The infant gut microbiota establishes during a critical window of opportunity when metabolic and immune functions are highly susceptible to environmental changes, such as diet. Human milk oligosaccharides (HMOs) for instance are suggested to be beneficial for infant health and gut microbiota. Infant formulas supplemented with the HMOs 2'-fucosyllactose (2'-FL) and lacto-N-neotetraose (LNnT) reduce infant morbidity and medication use and promote beneficial bacteria in the infant gut ecosystem. To further improve infant formula and achieve closer proximity to human milk composition, more complex HMO mixtures could be added. However, we currently lack knowledge about their effects on infants' gut ecosystems. METHOD: We assessed the effect of lactose, 2'-FL, 2'-FL + LNnT, and a mixture of six HMOs (HMO6: consisting of 2'-FL, LNnT, difucosyllactose, lacto-N-tetraose, 3'- and 6'-sialyllactose) on infant gut microbiota and intestinal barrier integrity using a combination of in vitro models to mimic the microbial ecosystem (baby M-SHIME®) and the intestinal epithelium (Caco-2/HT29-MTX co-culture). RESULTS: All the tested products had bifidogenic potential and increased SCFA levels; however, only the HMOs' fermented media protected against inflammatory intestinal barrier disruption. 2'-FL/LNnT and HMO6 promoted the highest diversification of OTUs within the Bifidobactericeae family, whereas beneficial butyrate-producers were specifically enriched by HMO6. CONCLUSION: These results suggest that increased complexity in HMO mixture composition may benefit the infant gut ecosystem, promoting different bifidobacterial communities and protecting the gut barrier against pro-inflammatory imbalances.


Subject(s)
Gastrointestinal Microbiome , Milk, Human , Caco-2 Cells , Ecosystem , Humans , Infant , Infant Formula , Lactose/metabolism , Lactose/pharmacology , Milk, Human/metabolism , Oligosaccharides/metabolism
2.
Nutrients ; 13(3)2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33668823

ABSTRACT

Because of the recognized health benefits of breast milk, it is recommended as the sole nutrition source during the first 6 months of life. Among the bioactive components are human milk oligosaccharides (HMOs) that exert part of their activity via the gut microbiota. Here, we investigated the gut microbiota fermentation of HMO 2'fucosyllactose (2'-FL), using two in vitro models (48 h fecal incubations and the long-term mucosal simulator of the human intestinal microbial ecosystem [M-SHIME®]) with fecal samples from 3-month-old breastfed (BF) infants as well as 2-3 year old toddlers. The short-term model allowed the screening of five donors for each group and provided supportive data for the M-SHIME® study. A key finding was the strong and immediate increase in the relative abundance of Bifidobacteriaceae following 2'-FL fermentation by both the BF infant and toddler microbiota in the M-SHIME®. At the metabolic level, while decreasing branched-chain fatty acids, 2'-FL strongly increased acetate production together with increases in the health-related propionate and butyrate whilst gas production only mildly increased. Notably, consistently lower gas production was observed with 2'-FL fermentation as compared to lactose, suggesting that reduced discomfort during the dynamic microbiome establishment in early life may be an advantage along with the bifidogenic effect observed.


Subject(s)
Gastrointestinal Microbiome/drug effects , Lactose/pharmacology , Milk, Human/chemistry , Trisaccharides/pharmacology , Bifidobacterium , Breast Feeding , Humans , Infant , Infant Nutritional Physiological Phenomena
3.
Sci Rep ; 10(1): 16117, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32999316

ABSTRACT

Numerous benefits of breastfeeding over infant formula are fully established. The superiority of human milk over bovine milk-based formula is partly due to human milk oligosaccharides (HMOs), a family of over 100 molecules present specifically and substantially in human milk that resemble mucosal glycans. To uncover novel physiological functions and pathways of HMOs, we screened a panel of 165 G-protein coupled receptors (GPCRs) using a blend of 6 HMOs (3'-O-sialyllactose (3'SL), 6'-O-sialyllactose (6'SL), lacto-N-tetraose (LNT), lacto-N-neo-tetraose (LNnT), 2-O-fucosyllactose (2'FL), and difucosyllactose (diFL)), and followed up positive hits with standard receptor assays. The HMO blend specifically activated GPR35. LNT and 6'SL individually activated GPR35, and they showed synergy when used together. In addition, in vitro fermentation of infant stool samples showed that 2'FL upregulates the production of the GPR35 agonist kynurenic acid (KYNA) by the microbiota. LNT + 6'SL and KYNA showed additive activation of GPR35. Activation by 6'SL and LNT of GPR35, a receptor mediating attenuation of pain and colitis, is to our knowledge the first demonstration of GPCR activation by any HMO. In addition, we demonstrated a remarkable cooperation between nutrition and microbiota towards activation of a host receptor highlighting the close interplay between environment and host-microbe interactions.


Subject(s)
Milk, Human/metabolism , Oligosaccharides/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Animals , Breast Feeding/methods , Cattle , GTP-Binding Proteins/metabolism , Host Microbial Interactions/physiology , Humans , Infant , Infant Formula , Lactose/analogs & derivatives , Lactose/metabolism , Trisaccharides/metabolism
4.
Environ Microbiol ; 16(9): 2891-904, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24033881

ABSTRACT

Breast milk has recently been recognized as source of commensal and potential probiotic bacteria. The present study investigated whether viable strains of gut-associated obligate anaerobes are shared between the maternal and neonatal gut ecosystem via breastfeeding. Maternal faeces, breast milk and corresponding neonatal faeces collected from seven mothers-neonate pairs at three neonatal sampling points were analyzed by culture-independent (pyrosequencing) and culture-dependent methods (16S rRNA gene sequencing, pulsed field gel electrophoresis, random amplified polymorphic DNA and repetitive extragenic palindromic polymerase chain reaction. Pyrosequencing allowed identifying gut-associated obligate anaerobic genera, like Bifidobacterium, Bacteroides, Parabacteroides and members of the Clostridia (Blautia, Clostridium, Collinsella and Veillonella) shared between maternal faeces, breast milk and neonatal faeces. Using culture, a viable strain of Bifidobacterium breve was shown to be shared between all three ecosystems within one mother-neonate pair. Furthermore, pyrosequencing revealed that several butyrate-producing members of the Clostridia (Coprococcus, Faecalibacterium, Roseburia and Subdoligranulum) were shared between maternal faeces and breast milk. This study shows that (viable) obligate gut-associated anaerobes may be vertically transferred from mother to neonate via breastfeeding. Thus, our data support the recently suggested hypothesis of a novel way of mother-neonate communication, in which maternal gut bacteria reach breast milk via an entero-mammary pathway to influence neonatal gut colonization and maturation of the immune system.


Subject(s)
Bacteria/classification , Breast Feeding , Feces/microbiology , Gastrointestinal Tract/microbiology , Milk, Human/microbiology , Bacteria/genetics , Bacteria, Anaerobic/classification , Bacteria, Anaerobic/genetics , Bifidobacterium/classification , Bifidobacterium/genetics , DNA Fingerprinting , DNA, Bacterial/genetics , Female , Gram-Positive Bacteria/classification , Gram-Positive Bacteria/genetics , Humans , Infant, Newborn , Probiotics , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
5.
J Pediatr Gastroenterol Nutr ; 52(6): 756-62, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21593648

ABSTRACT

OBJECTIVES: Breast-feeding induces a gut microbiota rich in bifidobacteria, whereas formula-fed babies have a more diverse colonization. This ecosystem contributes to the development of the immune response and the lower incidence of diarrhea and allergy in breast-fed infants. This randomized double-blind controlled trial aimed to evaluate the bifidogenic effect of a mainly whey protein study formula low in phosphate and protein, allowing a composition closer to that of human milk. PATIENTS AND METHODS: One hundred ninety healthy infants exclusively received study formula with or without Bifidobacterium longum (BL999), or a control formula for up to 4 months. Breast-fed infants served as a reference population. Stool samples collected at 2 months of age were analyzed for bacterial counts (log colony-forming unit [CFU]/g). RESULTS: Bifidobacteria counts were significantly higher in infants receiving the study formula alone (10.0[0.8], P < 0.0001, median [interquartile range]) or with BL999 (9.8[1.4], P < 0.01) than control (9.2[3.5]), and were similar to breast-fed infants (10.1[0.4], P > 0.05). The difference between the 2 study groups was 0.16 log CFU/g (90% confidence interval [CI] [0-0.4]), within the predefined equivalence margin. Microbiota profile, as a percentage of total bacteria counts, showed about 50% Bifidobacteria, 8% Enterobacteria, and <10% Clostridia in study formulae and breast-fed infants versus 22%, 13%, and 19% in controls, respectively. There were no significant differences in growth measurements, digestive tolerance, and adverse events between groups. CONCLUSIONS: This study showed that infant formula closer resembling human milk was more bifidogenic than the control formula and led to a microbiota profile similar to that for breast-fed infants.


Subject(s)
Bifidobacterium , Food Microbiology , Gastrointestinal Tract/microbiology , Infant Formula , Infant, Newborn , Metagenome , Probiotics , Bacterial Load , Breast Feeding , Clostridium/isolation & purification , Double-Blind Method , Enterobacteriaceae/isolation & purification , Feces/microbiology , Female , Humans , Male , Milk, Human/microbiology
6.
Anaerobe ; 16(4): 362-70, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20541022

ABSTRACT

Modifications in microbial colonization of the human gut are believed to affect intestinal homeostasis and increase the risk of gastrointestinal diseases. The present study examined different methods for investigating the dynamic characterization of the intestinal microbiota in preterm infants. Fecal samples were collected weekly from ten preterm infants during their stay in a neonatal intensive care unit. The infants had a mean gestational age of 29 weeks (range: 28-32 weeks) and a mean birth weight of 1233g (range: 935-1450g). Bacterial colonization was assessed using conventional culture techniques and molecular biological methods. More specifically, the recently developed denaturing high performance liquid chromatography (dHPLC) technique was compared to established methods such as temporal temperature gradient gel electrophoresis (TTGE) and rRNA gene library sequencing. Our results indicate that the gastrointestinal tract of preterm infants, born at a gestational age of less than 33 weeks, has a low biodiversity of mainly, culturable bacteria. Finally, dHPLC was evaluated in terms of speed, labor and sensitivity for its use as a tool to analyze microbial colonization in preterm infants. We found that this technique provided major improvements over gel-based fingerprinting methods, such as TTGE, that are commonly used for studying microbial ecology. As such, it may become a common analytical tool for this purpose.


Subject(s)
Biodiversity , Gastrointestinal Tract/microbiology , Metagenome , Premature Birth , Birth Weight , DNA Fingerprinting/methods , Electrophoresis, Polyacrylamide Gel , Feces/microbiology , Humans , Infant, Newborn , Intensive Care Units, Neonatal , Nucleic Acid Denaturation , Sequence Analysis, DNA/methods
7.
Ann Nutr Metab ; 55(4): 334-40, 2009.
Article in English | MEDLINE | ID: mdl-19844090

ABSTRACT

BACKGROUND/AIMS: Breast milk is the best source of nutrition for the growth of the newborn infant. It is therefore essential that mothers who cannot breastfeed or choose not to are provided with alternatives that closely match the composition and functionality of breast milk. This study aimed to investigate the growth effects of probiotic-supplemented formulas on both healthy and vulnerable populations of infants. METHODS: A meta-analysis of data from 5 randomized controlled clinical trials that included infants fed formulas containing a probiotic Bifidobacterium lactis CNCM I-3446 was performed (n = 525). A sub-analysis was performed among infants of HIV-positive mothers (n = 120). Growth measurements (gain in weight and body mass index, BMI, from enrollment to 120 days) were compared between infants fed a formula containing B. lactis and those fed a control formula. Changes in length and Z-scores were also compared. RESULTS: Formula with B.lactis was demonstrated to be at least as good as formula without B. lactis in the meta-analysis of 5 studies. The lower boundary of the 95% confidence interval (CI) of the differences in mean weight gain (95% CI 0.09-2.93 g/day) was above the predefined non-inferiority margin of -3.0 g/day. Moreover, among infants with HIV-positive mothers, weight gain of those taking B. lactis was significantly higher than of those not taking B. lactis, by 3.1 g/day (95% CI 0.4-5.8 g/day, p = 0.0226) and the BMI gains were significantly higher, by 6.4 g/m(2)/day (95% CI 0.0.3-12.5 g/m(2)/day, p = 0.0400). The corresponding weight for age and BMI Z-scores were also significantly higher, by 0.37 (95% CI 0.03-0.71, p = 0.0308) and by 0.42 (95% CI 0.02-0.83, p = 0.0377), respectively, whereas differences in length gain or length-for-age Z-score were not significant. Among infants in the non-HIV mothers group, there were no significant differences between infants fed formulas with or without B. lactis, for any of the growth parameters. CONCLUSIONS: The analysis suggests that B. lactis may have a positive effect on growth in vulnerable populations, specifically in infants born to mothers with HIV.


Subject(s)
Bifidobacterium , Infant Formula , Probiotics , Weight Gain , Anthropometry , Female , HIV Infections , Humans , Infant , Infant, Newborn , Male , Mothers , Randomized Controlled Trials as Topic
8.
Am J Clin Nutr ; 89(6): 1828-35, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19369375

ABSTRACT

BACKGROUND: Although recent reports suggest that supplementation with probiotics may enhance intestinal function in premature infants, the mechanisms are unclear, and questions remain regarding the safety and efficacy of probiotics in extremely low-birth-weight infants. OBJECTIVE: The objective was to evaluate the efficacy of probiotics on the digestive tolerance to enteral feeding in preterm infants born with a very low or extremely low birth weight. DESIGN: In a bicentric, double-blind, randomized controlled clinical trial that was stratified for center and birth weight, 45 infants received enteral probiotics (Bifidobacterium longum BB536 and Lactobacillus rhamnosus GG; BB536-LGG) and 49 received placebo. The primary endpoint was the percentage of infants receiving >50% of their nutritional needs via enteral feeding on the 14th day of life. A triangular test was used to perform sequential analysis. RESULTS: The trial was discontinued after the fourth sequential analysis concluded a lack of effect. The primary endpoint was not significantly different between the probiotic (57.8%) and placebo (57.1%) groups (P = 0.95). However, in infants who weighed >1000 g, probiotic supplementation was associated with a shortening in the time to reach full enteral feeding (P = 0.04). Other than colonization by the probiotic strains, no alteration in the composition of intestinal microbiota or changes in the fecal excretion of calprotectin was observed. No colonization by probiotic strains was detected in infants who weighed < or =1000 g, presumably because of more frequent suspensions of enteral feeding, more courses of antibiotic treatment, or both. CONCLUSIONS: Supplementation with BB536-LGG may not improve the gastrointestinal tolerance to enteral feeding in very-low-birth-weight infants but may improve gastrointestinal tolerance in infants weighing >1000 g. This trial was registered at clinicaltrials.gov as NCT 00290576.


Subject(s)
Bifidobacterium , Enteral Nutrition , Infant, Premature/growth & development , Infant, Very Low Birth Weight/growth & development , Intestines/microbiology , Lacticaseibacillus rhamnosus , Probiotics/pharmacology , Dietary Supplements , Double-Blind Method , Humans , Infant , Infant, Extremely Low Birth Weight/growth & development , Infant, Newborn , Leukocyte L1 Antigen Complex/analysis , Probiotics/administration & dosage , Treatment Outcome , Weight Gain/drug effects
9.
J Proteome Res ; 8(4): 2090-105, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19281268

ABSTRACT

Coevolution shapes interorganismal crosstalk leading to profound and diverse cellular and metabolic changes as observed in gut dysbiosis in human diseases. Here, we modulated a simplified gut microbiota using pro-, pre-, and synbiotics to assess the depth of systemic metabolic exchanges in mice, using a multicompartmental modeling approach with metabolic signatures from 10 tissue/fluid compartments. The nutritionally induced microbial changes modulated host lipid, carbohydrate, and amino acid metabolism at a panorganismal scale. Galactosyl-oligosaccharides reduced lipogenesis, triacylglycerol incorporation into lipoproteins and triglyceride concentration in the liver and the kidney. Those changes were not correlated with decreased plasma lipoproteins that were specifically induced by L. rhamnosus supplementation. Additional alteration of transmethylation metabolic pathways (homocysteine-betaine) was observed in the liver and the pancreas following pre- and synbiotic microbial modulation, which may be of interest for control of glucose metabolism and insulin sensitivity. Probiotics also reduced hepatic glycogen and glutamine and adrenal ascorbate with inferred effects on energy homeostasis, antioxidation, and steroidogenesis. These studies show the breadth and the depth of gut microbiome modulations of host biochemistry and reveal that major mammalian metabolic processes are under symbiotic homeostatic control.


Subject(s)
Gastrointestinal Tract/microbiology , Metabolome , Metagenome , Animals , Female , Gastrointestinal Tract/metabolism , Humans , Liver/metabolism , Liver/microbiology , Mice , Nuclear Magnetic Resonance, Biomolecular , Probiotics/metabolism
10.
Mol Syst Biol ; 4: 205, 2008.
Article in English | MEDLINE | ID: mdl-18628745

ABSTRACT

Gut microbiome-host metabolic interactions affect human health and can be modified by probiotic and prebiotic supplementation. Here, we have assessed the effects of consumption of a combination of probiotics (Lactobacillus paracasei or L. rhamnosus) and two galactosyl-oligosaccharide prebiotics on the symbiotic microbiome-mammalian supersystem using integrative metabolic profiling and modeling of multiple compartments in germ-free mice inoculated with a model of human baby microbiota. We have shown specific impacts of two prebiotics on the microbial populations of HBM mice when co-administered with two probiotics. We observed an increase in the populations of Bifidobacterium longum and B. breve, and a reduction in Clostridium perfringens, which were more marked when combining prebiotics with L. rhamnosus. In turn, these microbial effects were associated with modulation of a range of host metabolic pathways observed via changes in lipid profiles, gluconeogenesis, and amino-acid and methylamine metabolism associated to fermentation of carbohydrates by different bacterial strains. These results provide evidence for the potential use of prebiotics for beneficially modifying the gut microbial balance as well as host energy and lipid homeostasis.


Subject(s)
Genome/genetics , Intestines/microbiology , Lactobacillus/genetics , Lactobacillus/metabolism , Models, Animal , Probiotics , Systems Biology , Animals , Body Weight , Cecum/metabolism , Fatty Acids/metabolism , Feces/microbiology , Female , Genome/drug effects , Humans , Infant , Intestines/drug effects , Liver/metabolism , Magnetic Resonance Spectroscopy , Mice , Probiotics/pharmacology
11.
Br J Nutr ; 100(5): 1077-85, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18377682

ABSTRACT

Specific carbohydrates, i.e. prebiotics such as fructo-oligosaccharide (FOS), are not digested in the small intestine but fermented in the colon. Besides beneficial health effects of an enhanced bifidobacteria population, intestinal gas production resulting from fermentation can induce abdominal symptoms. Partial replacement with slowly fermented acacia gum may attenuate side effects. The aim was to compare the effects of FOS with those of a prebiotic mixture (50 % FOS and 50 % acacia gum; BLEND) and a rapidly absorbed carbohydrate (maltodextrin) on general intestinal wellbeing, abdominal comfort and anorectal sensory function. Twenty volunteers (eight male and twelve female; age 20-37 years) completed this double-blind, randomised study with two cycles of a 2-week run-in phase (10 g maltodextrin) followed by 5 weeks of 10 g FOS or BLEND once daily, separated by a 4-week wash-out interval. Abdominal symptoms and general wellbeing were documented by telephone interview or Internet twice weekly. Rectal sensations were assessed by a visual analogue scale during a rectal barostat test after FOS and BLEND treatment. Both FOS and BLEND induced more side effects than maltodextrin. Belching was more pronounced under FOS compared with BLEND (P = 0.09 for females; P = 0.01 for males), and for self-reported general wellbeing strong sex differences were reported (P = 0.002). Urgency scores during rectal barostat were higher with FOS than BLEND (P = 0.01). Faced with a growing range of supplemented food products, consumers may benefit from prebiotic mixtures which cause fewer abdominal side effects. Sex differences must be taken in consideration when food supplements are used.


Subject(s)
Dietary Carbohydrates/administration & dosage , Gum Arabic/administration & dosage , Health , Intestines/microbiology , Oligosaccharides/administration & dosage , Polysaccharides/administration & dosage , Adult , Bifidobacterium/metabolism , Colon/microbiology , Cross-Over Studies , Dietary Supplements , Double-Blind Method , Female , Fermentation , Flatulence , Humans , Male , Probiotics/metabolism , Sex Factors , Surveys and Questionnaires , Telemedicine/methods
12.
J Nutr ; 138(1): 123-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18156414

ABSTRACT

Fructooligosaccharides (FOS) are considered prebiotics because of their ability to promote growth of specific beneficial gut bacteria, such as bifidobacteria. Some studies reported potential immune-modulating properties. The aim of this study was to investigate the effect of FOS:inulin mix on murine response to Salmonella vaccine and evaluate the relevance toward protection against Salmonella infection. Balb/c mice were fed a diet containing 5% FOS:inulin mix or a control diet 1 wk before oral immunization with a suboptimal dose of live attenuated Salmonella typhimurium vaccine. Four weeks after vaccination, mice were infected with LD100 of virulent S. typhimurium. Specific blood Salmonella immunoglobulin G and fecal immunoglobulin A significantly increased in mice fed the diet containing prebiotics compared with control mice 4 wk postimmunization. Peritoneal macrophage phagocytic activity also significantly increased in FOS:inulin-fed mice at 1 wk postimmunization compared with control mice. No detectable effects were observed on the percentage of lymphoid cell subsets in the spleen. However, production of cytokines, interferon-gamma, interleukin-12, and tumor necrosis factor alpha, was numerically increased in spleen cell cultures stimulated with mitogens from FOS:inulin-fed mice 1 and 4 wk postimmunization. Salmonella translocation to lymphoid organs was not affected by feeding FOS:inulin. However, the improved response to Salmonella vaccine was concomitant with an increase in the survival rate of FOS:inulin-fed mice upon challenge with virulent Salmonella. No detectable effects were observed on the composition or the metabolic activity of the microbiota. Overall, the data suggest that a diet supplemented with FOS:inulin mix stimulates mucosal immunity and seems to improve efficacy of an oral vaccine.


Subject(s)
Diet , Inulin/administration & dosage , Oligosaccharides/administration & dosage , Salmonella Vaccines/immunology , Administration, Oral , Animals , Antibodies, Bacterial , Cytokines/metabolism , Female , Immunoglobulin A/metabolism , Immunoglobulin G/blood , Inulin/pharmacology , Metalloporphyrins , Mice , Mice, Inbred BALB C , Oligosaccharides/pharmacology , Phagocytes , Salmonella Infections/prevention & control , Salmonella Vaccines/administration & dosage , Spleen/cytology , Spleen/metabolism
13.
Genomics ; 91(1): 70-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18035521

ABSTRACT

The intestinal mucus layer and endogenous microbiota are strongly intertwined and this contributes to the maintenance of the epithelial barrier and ultimately of gut homeostasis. To understand the molecular foundations of such relationship, we investigated if the nature of the microbiota transcriptionally regulates mucus layer composition in vivo. We found that the expression of mucins 1 to 4 and trefoil factor 3 was down-regulated in the ileum and colon of conventional and reconventionalized mice compared with germ-free animals. Conversely, very limited colon-restricted changes in transmembrane mucins were detected in mice colonized with human adult or baby microbiota. Moreover, by microarray analysis, the murine endogenous microbiota was found to modulate genes putatively involved in mucin secretion. These findings show that a well-established microbial community participates in the regulation of the gut mucus layer and that its composition and adequacy to the host are key factors in this process.


Subject(s)
Down-Regulation/physiology , Germ-Free Life/physiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mucins/biosynthesis , Adult , Animals , Colon/metabolism , Colon/microbiology , Gene Expression Profiling , Humans , Ileum/metabolism , Ileum/microbiology , Infant , Infant, Newborn , Male , Mice , Mice, Inbred C3H , Mucins/genetics , Oligonucleotide Array Sequence Analysis , Trefoil Factor-3
14.
Br J Nutr ; 99(2): 287-96, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17761014

ABSTRACT

Chicory roots are rich in inulin that is degraded into SCFA in the caecum and colon. Whole-body SCFA metabolism was investigated in rats during food deprivation and postprandial states. After 22 h of food deprivation, sixteen rats received an IV injection of radioactive 14C-labelled SCFA. The volume of distribution and the fractional clearance rate of SCFA were 0.25-0.27 litres/kg and 5.4-5.9 %/min, respectively. The half-life in the first extracellular rapidly decaying compartment was between 0.9 and 1.4 min. After 22 h of food deprivation, another seventeen rats received a primed continuous IV infusion of 13C-labelled SCFA for 2 h. Isotope enrichment (13C) of SCFA was determined in peripheral arterial blood by MS. Peripheral acetate, propionate and butyrate turnover rates were 29, 4 and 0.3 micromol/kg per min respectively. Following 4 weeks of treatment with chicory root or control diets, eighteen fed rats received a primed continuous IV infusion of 13C-labelled SCFA for 2 h. Intestinal degradation of dietary chicory lowered caecal pH, enhanced caecal and colonic weights, caecal SCFA concentrations and breath H2. The diet with chicory supplementation enhanced peripheral acetate turnover by 25 % (P = 0.017) concomitant with an increase in plasma acetate concentration. There were no changes in propionate or butyrate turnovers. In conclusion, by setting up a multi-tracer approach to simultaneously assess the turnovers of acetate, propionate and butyrate it was demonstrated that a chronic chicory-rich diet significantly increases peripheral acetate turnover but not that of propionate or butyrate in rats.


Subject(s)
Acetates/blood , Cichorium intybus , Fatty Acids, Volatile/blood , Animals , Butyrates/blood , Carbon Radioisotopes , Cecum/metabolism , Colon/metabolism , Fatty Acids, Volatile/biosynthesis , Female , Food Deprivation/physiology , Half-Life , Male , Portal Vein/metabolism , Postprandial Period/physiology , Propionates/blood , Rats
15.
J Nutr ; 137(8): 1901-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17634262

ABSTRACT

Stressful events result in the alteration of gut permeability and sensitivity. Lactobacillus paracasei NCC2461 (Lpa) therapy prevents antibiotic-induced visceral hyperalgesia in mice. This study aimed at evaluating the influence of 3 probiotic strains: Bifidobacterium lactis NCC362, Lactobacillus johnsonii NCC533, and Lpa on stress-mediated alterations of colorectal hyperalgesia, on gut paracellular permeability and whether bacteria and/or bacterial products present in the spent culture medium (SCM) were involved in the antinociceptive properties of the effective strain. Rat pups were separated from their mothers 3 h/d during postnatal d 2-14. At wk 13, gut paracellular permeability was determined as a percentage of urinary excreted (51)Cr-EDTA and visceral sensitivity to colorectal distension (CRD), assessed by abdominal muscle electromyography. Visceral sensitivity was also analyzed in adults rats subjected to partial restraint stress (PRS, 2 h restriction of body movements). Rats received either the probiotics resuspended in SCM or fresh growth medium as control for 2 wk. Maternal deprivation significantly increased colonic sensitivity in response to CRD and enhanced gut paracellular permeability compared with control rats. Only Lpa treatment significantly improved stress-induced visceral pain and restored normal gut permeability. Similarly, among the 3 probiotics tested, only Lpa prevented PRS-mediated visceral hyperalgesia. Both bacteria and bacterial products present in Lpa SCM were required for the antinociceptive properties against PRS. This study illustrates strain-specific effects and suggests a synergistic interplay between L. paracasei bacteria and bacterial products generated during fermentation and growth that confers the ability to suppress PRS-induced hypersensitivity in rats.


Subject(s)
Intestines/microbiology , Intestines/physiology , Lactobacillus/metabolism , Permeability , Stress, Physiological/metabolism , Animals , Bifidobacterium/metabolism , Pain , Rats , Rats, Wistar , Restraint, Physical
16.
Nutrition ; 23(1): 1-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17189085

ABSTRACT

OBJECTIVES: The larger number of bifidobacteria in the intestine of breast-fed infants has been associated with their better health compared with formula-fed infants. We assessed the safety and tolerability of an experimental formula containing 2 x 10(7) colony-forming units of Bifidobacterium longum BL999 and 4 g/L of a prebiotic mixture containing 90% galacto-oligosaccharides and 10% fructo-oligosaccharides. METHODS: A 7-mo prospective, randomized, reference-controlled, double-blinded trial was performed in infants who were not breast fed after the 14th day of birth. One hundred thirty-eight infants were enrolled and assigned to receive the control or experimental formula until they were 112 d old. Mean weight gain (primary outcome) and recumbent length, head circumference, tolerability (gastrointestinal symptoms), and overall morbidity (secondary outcomes) were measured at 14, 28, 56, 84, and 112 d of age. RESULTS: Equivalence in mean weight gain between the two groups was shown. The treatment difference in the intention-to-treat and per-protocol populations were within the predefined equivalence boundaries of +/-3.9 g/d. No statistically significant difference in recumbent length, head circumference, or incidence of adverse events was found between the two groups. Infants in the experimental group had fewer incidences of constipation and had stool characteristics that suggest that the experimental formula was tolerated well. Furthermore, these infants showed a trend toward fewer respiratory tract infections. CONCLUSIONS: The starter formula containing BL999 and galacto-oligosaccharides/fructo-oligosaccharides is safe and well-tolerated.


Subject(s)
Bifidobacterium/physiology , Infant Nutritional Physiological Phenomena , Probiotics/administration & dosage , Probiotics/adverse effects , Weight Gain , Bifidobacterium/growth & development , Body Height , Colony Count, Microbial , Constipation/epidemiology , Consumer Product Safety , Double-Blind Method , Female , Head/anatomy & histology , Humans , Infant , Infant Formula , Infant, Newborn , Intestines/microbiology , Male , Oligosaccharides/administration & dosage , Oligosaccharides/adverse effects , Prospective Studies , Respiratory Tract Infections/epidemiology
17.
J Pediatr Gastroenterol Nutr ; 43(1): 16-24, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16819372

ABSTRACT

OBJECTIVES: Psychological stress during the neonatal period results in intestinal barrier dysfunction and growth alterations later in life. We aimed to restore impaired barrier function and growth rate by a nutritional intervention. METHODS: Male rat pups (n = 84) were assigned to 1 of 2 rearing conditions from postnatal day (PND) 2 to PND14: S, separated 3 h/d from their mothers, or H, 15 min/d handled controls. From PND15 to PND35, rats received a control diet or a similar diet adapted to contain arachidonic and docosahexaenoic acids, galacto- and fructo-oligosaccharides and Lactobacillus paracasei NCC2461. RESULTS: Maternal separation had only a minor impact on the measured gut barrier parameters at PND15, whereas it severely affected them at PND35. At this age, intestinal permeability to macromolecules was higher, mucin content in small intestinal tissues was lower and microbiota composition was altered in S compared with H animals. Feeding the adapted diet normalized the intestinal permeability, although it did not restore intestinal mucin content or microbiota. In addition, the adapted diet improved the growth rate recovery of the S animals after weaning and resulted in increased villus length in small intestine. CONCLUSION: Our results suggest that an adapted diet containing specific long-chain polyunsaturated fatty acids, prebiotics and probiotics can revert the negative imprinting of neonatal stress on both intestinal barrier function and growth.


Subject(s)
Food, Formulated , Intestinal Mucosa/growth & development , Intestinal Mucosa/physiopathology , Intestine, Small/growth & development , Intestine, Small/physiopathology , Stress, Psychological/physiopathology , Animals , Animals, Newborn , Arachidonic Acid/administration & dosage , Arachidonic Acid/metabolism , Body Weight , Disease Models, Animal , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/metabolism , Eating , Homeostasis , Intestinal Absorption , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestine, Small/metabolism , Intestine, Small/microbiology , Lactobacillus/metabolism , Male , Maternal Deprivation , Mucins/metabolism , Oligosaccharides/administration & dosage , Oligosaccharides/metabolism , Permeability , Probiotics/administration & dosage , Probiotics/metabolism , Rats , Rats, Long-Evans , Stress, Psychological/metabolism , Stress, Psychological/microbiology , Time Factors , Weaning
18.
Asia Pac J Clin Nutr ; 15(3): 368-76, 2006.
Article in English | MEDLINE | ID: mdl-16837430

ABSTRACT

The aim of the study was to evaluate whether supplementation of milk-formulas with prebiotic fructo-oligosaccharides or a probiotic, Lactobacillus johnsonii La1 (La1), could modulate the composition of the fecal microbiota of formula-fed infants, compared to breastfed (BF) infants. Ninety infants close to 4 months of age were randomized into one of three groups to be blindly assigned to receive for 13 weeks: a) an infant formula (Control), b) the same formula with fructo-oligosaccharides (Prebio), or c) with La1 (Probio). At the end of this period, all infants received the control formula for 2 additional weeks. Twenty-six infants, breastfed throughout the study, were recruited to form group BF. Fecal samples were obtained upon enrolment and after 7 and 15 weeks. Bacterial populations were assessed with classical culture techniques and fluorescent in situ hybridisation (FISH). Seventy-six infants completed the study. On enrolment, higher counts of Bifidobacterium and Lactobacillus and lower counts of enterobacteria were observed in BF compared to the formula-fed infants; these differences tended to disappear at weeks 7 and 15. No major differences for Clostridium, Bacteroides or Enterococcus were observed between the groups or along the follow up. Probio increased fecal Lactobacillus counts (p<0.001); 88% of the infants in this group excreted live La1 in their stools at week 7 but only 17% at week 15. Increased Bifidobacterium counts were observed at week 7 in the 3 formula groups, similar to BF infants. These results confirm the presence of higher counts of bifidobacteria and lactobacilli in the microbiota of BF infants compared to formula-fed infants before dietary diversification, and that La1 survives in the infant digestive tract.


Subject(s)
Feces/microbiology , Infant Formula , Oligosaccharides/administration & dosage , Probiotics/administration & dosage , Bifidobacterium , Breast Feeding , Colony Count, Microbial , Enterobacteriaceae , Female , Humans , Infant, Newborn , Lactobacillus , Male , Prospective Studies
19.
J Nutr ; 136(6): 1558-64, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16702321

ABSTRACT

During the anabolic response associated with inflammation, mucin synthesis and colonic protection may be compromised by the limited availability of specific amino acids. We therefore determined the effect of dietary amino acid supplementation on the microbiota, mucin status, and mucosal damage in dextran sulfate sodium (DSS)-treated rats. From 8 d before to 28 d after colitis induction, male Sprague-Dawley rats (10 mo old, n = 8/group) were fed a control diet supplemented or not with 2 different doses of an amino acid cocktail containing L-threonine, L-serine, L-proline, and L-cysteine. All diets were isonitrogenous (adjusted with L-alanine). The higher dose of amino acids increased the number of Muc2-containing goblet cells in the surface epithelium of the ulcerated area, stimulated mucin production in the colon, and restored the mucin amino acid composition and mucosal content to healthy, control values. The colonic mucin synthesis rate was specifically stimulated by 95%, whereas the protein turnover was unchanged. All bacterial populations, markedly altered by the DSS treatment, were promoted. In conclusion, in inflammatory situations, an increase in threonine, serine, proline, and cysteine dietary supply can promote mucin synthesis, reequilibrate the gut microbiota, and thus favor colonic protection and mucosal healing.


Subject(s)
Amino Acids/therapeutic use , Anticoagulants/pharmacology , Dextran Sulfate/pharmacology , Goblet Cells/pathology , Intestines/drug effects , Mucins/biosynthesis , Proteins/metabolism , Amino Acids/administration & dosage , Animals , Body Weight/drug effects , Colitis, Ulcerative/prevention & control , Disease Models, Animal , Feces/microbiology , Goblet Cells/drug effects , Intestinal Mucosa/metabolism , Male , Mucin-2 , Mucins/genetics , Organ Size/drug effects , Rats , Rats, Sprague-Dawley
20.
Br J Nutr ; 95(2): 303-12, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16469146

ABSTRACT

To exert beneficial effects for the host, for example, improving the intestinal microflora, a probiotic must reach the intestine as a viable strain. These properties must be demonstrated by in vitro as well as in vivo methods. However, only a few well-designed human clinical studies have shown these properties. Lactobacillus johnsonii La1 has been shown to give many beneficial effects for the host, but it is unclear whether a viable strain of L. johnsonii La1 has the effect of improving host intestinal microflora. In the present study, a randomised double-blind placebo-controlled cross-over trial was conducted to elucidate the effect of L. johnsonii La1 on human intestinal microflora. Twenty-two young healthy Japanese women were randomly divided into two groups, and either received fermented milk with L. johnsonii La1 or a fermented milk without L. johnsonii La1 (placebo) daily for 21 d. Consumption of the fermented milk: (a) increased total Bifidobacterium and Lactobacillus, and decreased lecithinase-positive Clostridium in the faeces; (b) increased the faecal lactic acid concentrations; (c) decreased the faecal pH; (d) increased the defecation frequency. These changes were stronger than those observed with the placebo. L. johnsonii La1 was identified in all subjects only after the consumption of the fermented milk. These results suggest that L. johnsonii La1 can contribute to improve intestinal microflora with probiotic properties.


Subject(s)
Intestines/microbiology , Lactobacillus , Probiotics/administration & dosage , Adult , Bacterial Adhesion/physiology , Bifidobacterium/growth & development , Bifidobacterium/isolation & purification , Caco-2 Cells , Clostridium/isolation & purification , Cross-Over Studies , Culture Media , Cultured Milk Products , Defecation/physiology , Double-Blind Method , Feces/chemistry , Feces/microbiology , Female , Humans , Hydrogen-Ion Concentration , Lactic Acid/analysis , Lactobacillus/growth & development , Lactobacillus/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...