Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Nutrients ; 12(5)2020 May 20.
Article in English | MEDLINE | ID: mdl-32443684

ABSTRACT

BACKGROUND: Post-natal gut maturation in infants interrelates maturation of the morphology, digestive, and immunological functions and gut microbiota development. Here, we explored both microbiota development and markers of gut barrier and maturation in healthy term infants during their early life to assess the interconnection of gut functions during different infant formulae regimes. METHODS: A total of 203 infants were enrolled in this randomized double-blind controlled trial including a breastfed reference group. Infants were fed starter formulae for the first four weeks of life, supplemented with different combination of nutrients (lactoferrin, probiotics (Bifidobacterium animal subsp. Lactis) and prebiotics (Bovine Milk-derived Oligosaccharides-BMOS)) and subsequently fed the control formula up to eight weeks of life. Stool microbiota profiles and biomarkers of early gut maturation, calprotectin (primary outcome), elastase, α-1 antitrypsin (AAT) and neopterin were measured in feces at one, two, four, and eight weeks. RESULTS: Infants fed formula containing BMOS had lower mean calprotectin levels over the first two to four weeks compared to the other formula groups. Elastase and AAT levels were closer to levels observed in breastfed infants. No differences were observed for neopterin. Global differences between the bacterial communities of all groups were assessed by constrained multivariate analysis with hypothesis testing. The canonical correspondence analysis (CCA) at genus level showed overlap between microbiota profiles at one and four weeks of age in the BMOS supplemented formula group with the breastfed reference, dominated by bifidobacteria. Microbiota profiles of all groups at four weeks were significantly associated with the calprotectin levels at 4 (CCA, p = 0.018) and eight weeks of age (CCA, p = 0.026). CONCLUSION: A meaningful correlation was observed between changes in microbiota composition and gut maturation marker calprotectin. The supplementation with BMOS seems to favor gut maturation closer to that of breastfed infants.


Subject(s)
Biomarkers , Dietary Supplements , Gastrointestinal Microbiome/physiology , Infant Formula/analysis , Animals , Bifidobacterium animalis , Breast Feeding , Double-Blind Method , Feces/microbiology , Humans , Infant , Leukocyte L1 Antigen Complex , Milk , Oligosaccharides/analysis , Prebiotics/analysis , Probiotics/analysis
2.
Skin Pharmacol Physiol ; 32(4): 212-223, 2019.
Article in English | MEDLINE | ID: mdl-31167217

ABSTRACT

BACKGROUND: Little is known about the impact of nutrition on the development of skin structure and function in infants. METHODS: We investigated epidermal, dermal, and subcutis parameters of aged-matched well-nourished and moderately undernourished infants in this single-center, cross-sectional, noninterventional study using noninvasive methods (skin caliper, 20-MHz sonography, transepidermal water loss, skin pH, and corneometry). Plasma fatty acids were determined as an indicator of nutritional differences. 310 infants from different age groups, i.e., 1 week, 4 weeks, and 6, 9, 12, 24, and 36 months were included. Approximately half of each age group was well-nourished (WHO reference values weight-for-height/length Zscore: -0.75 ≤ Z ≤ 0.75) and the other half was moderately undernourished (-3 ≤ Z < -2). RESULTS: Structural maturational differences in the deeper dermis and subcutis regions of the skin and subtle functional changes in the epidermis were observed in moderately undernourished infants without notable clinical symptoms. Reduced skin barrier function or skin hydration were not observed in the undernourished infants, and skin pH shifted to more acidic values in this group. CONCLUSION: These findings reveal a greater impact of moderate undernutrition on the development of the dermis and subcutis and suggest that critical epidermal functions such as skin barrier and pH are mostly maintained.


Subject(s)
Infant Nutrition Disorders/physiopathology , Nutritional Status/physiology , Skin/physiopathology , Child, Preschool , Cross-Sectional Studies , Fatty Acids/metabolism , Female , Humans , Infant , Infant, Newborn , Male
3.
Int Arch Allergy Immunol ; 177(2): 123-134, 2018.
Article in English | MEDLINE | ID: mdl-30001534

ABSTRACT

Limited evidence is available regarding the effect of partially hydrolyzed whey-based formula (pHF-W) on growth and atopic dermatitis (AD) risk reduction in infants within the general infant population, and without a familial history of allergy as an inclusion or exclusion criterion. We reviewed the current evidence available from studies using pHF-W in the general population and summarized the data on safety (growth) and efficacy outcomes (reduction of AD), comparing the studies side by side. A total of 8 clinical trials were identified from the literature search, 7 of which used the same pHF-W. Six out of 8 studies indicated a reduction of atopic manifestations using a specific pHF-W versus cow's milk formula (CMF) in the first years of life. Data were summarized and compared side by side for growth (3 studies) and efficacy (5 studies). In these diverse general populations, the results on growth and AD were consistent with the previous findings reported on infants with a family history of allergy, but numerous limitations to these studies were identified. This literature review confirms that pHF-W supports normal growth in infants, and suggests that the risk of AD may be reduced in not-fully breastfed infants from the general population when supplemented with a specific pHF-W when compared to CMF during the first 4-6 months of life. Further studies are warranted to confirm these results.


Subject(s)
Dermatitis, Atopic/chemically induced , Infant Formula/chemistry , Whey Proteins/metabolism , Dermatitis, Atopic/physiopathology , Food Hypersensitivity/immunology , Humans , Hydrolysis , Infant , Infant, Newborn , Risk Factors
4.
BMC Pregnancy Childbirth ; 18(1): 193, 2018 May 31.
Article in English | MEDLINE | ID: mdl-29855271

ABSTRACT

BACKGROUND: Adequate nutrition is essential during pregnancy and lactation to provide sufficient energy and nutrients to meet the nutritional requirements of the mother, fetus and infant. The primary objective of this study was to assess the effect of a maternal nutritional supplement enriched with probiotics during pregnancy and early lactation on the incidence of infant diarrhea. METHODS: Healthy, pregnant (24-28 weeks gestation) women were randomized 1:1:1 to receive either no supplement or two servings per day of an oral supplement (140 kcal/serving) providing 7.9 g protein, multivitamin/minerals, and enriched or not with the probiotics Lactobacillus rhamnosus and Bifidobacterium lactis, from the third trimester of pregnancy until at least 2 months post-delivery. Incidence of infant diarrhea until 12 months post-delivery was analyzed by Poisson regression. The effect on maternal health, fetal growth, and infant growth and morbidity were also evaluated and analyzed by ANOVA. RESULTS: A total of 208 mother/infant pairs were included in the analysis. No significant difference in the incidence of infant diarrhea was observed between the three study groups. The mean maternal weight gains at delivery were similar among groups, despite an increase in caloric intake in the supplemented groups. No statistically significant differences between groups were observed in incidence of pregnancy-related or fetal adverse outcomes. Mean weight-, length-, BMI- and head circumference-for-age z-scores were below the WHO median value for all groups. Post-hoc analysis to compare the effect of the combined supplement groups versus the no supplement group on infant growth parameters showed, at 12 months, that the combined supplemented group had gained statistically significant more weight (8.97 vs. 8.61 kg, p = 0.001) and height (74.2 vs. 73.4 cm, p = 0.031), and had a higher weight-for-age z-score (- 0.62 vs. -0.88, p = 0.045) than the no supplement group. CONCLUSIONS: Maternal nutritional supplement with or without probiotics given during late pregnancy and early lactation was well tolerated and safe. Even though no difference in incidence of infant diarrhea was observed between the three groups, the analysis of the combined supplemented groups showed beneficial effects of maternal supplementation on infant weight and length gains at 12 months. TRIAL REGISTRATION: ClinicalTrial.gov: NCT01073033 . Registered 17.02.2010.


Subject(s)
Beverages , Dietary Supplements , Maternal Nutritional Physiological Phenomena , Prenatal Care/methods , Probiotics/administration & dosage , Adult , Birth Weight , Breast Feeding , Diarrhea/epidemiology , Female , Fetal Development , Humans , Incidence , Infant , Infant Health , Infant, Newborn , Infant, Newborn, Diseases/epidemiology , Lactation , Nutritional Status , Philippines/epidemiology , Poisson Distribution , Pregnancy , Pregnancy Trimester, Third , Regression Analysis , Young Adult
6.
Clin Med Insights Pediatr ; 11: 1179556517730018, 2017.
Article in English | MEDLINE | ID: mdl-28959122

ABSTRACT

To date, only few studies have assessed oral immunotherapy (OIT) for wheat allergy and often describe severe adverse reactions during therapy. We developed partially hydrolyzed wheat-based cereals (pHC), which were used in a multicenter, open-label, OIT pilot study, in immunoglobulin E-mediated wheat allergy children (NCT01332084). The primary objective of the study was to test whether wheat allergic patients tolerate pHC and primary end point was the presence or not of immediate adverse reactions to pHC during the 1-day initial escalation phase (stepwise increased doses of pHC), with evaluation of the maximum dose tolerated. Of the 9 patients enrolled in the trial, 4 discontinued OIT because of mild to severe reactions at the initial escalation phase. The 5 patients who passed the escalation phase consumed pHC daily for 1 to 6 months. One of these patients withdrew due to noncompliance, whereas the 4 others completed the study and successfully passed the wheat challenge test at the end of the study. About 60% of the adverse events were unrelated to the study product. Our study provides preliminary evidence that pHC is tolerated by a subset of wheat allergic patients. Further studies are warranted to test its efficacy as a potential therapeutic option for wheat allergic patients.

7.
Nutrients ; 9(3)2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28257044

ABSTRACT

The nutritional composition of human milk evolves over the course of lactation, to match the changing needs of infants. This single-arm, non-inferiority study evaluated growth against the WHO standards in the first year of life, in infants consecutively fed four age-based formulas with compositions tailored to infants' nutritional needs during the 1st, 2nd, 3rd-6th, and 7th-12th months of age. Healthy full-term formula-fed infants (n = 32) were enrolled at ≤14 days of age and exclusively fed study formulas from enrollment, to the age of four months. Powdered study formulas were provided in single-serving capsules that were reconstituted using a dedicated automated preparation system, to ensure precise, hygienic preparation. The primary outcome was the weight-for-age z-score (WAZ) at the age of four months (vs. non-inferiority margin of -0.5 SD). Mean (95% CI) z-scores for the WAZ (0.12 (-0.15, 0.39)), as well as for the length-for-age (0.05 (-0.19, 0.30)), weight-for-length (0.16 (-0.16, 0.48)), BMI-for-age (0.11 (-0.20, 0.43)), and head circumferencefor-age (0.41 (0.16, 0.65)) at the age of four months, were non-inferior. Throughout the study, anthropometric z-scores tracked closely against the WHO standards (within ±1 SD). In sum, a fourstage, age-based infant formula system with nutritional compositions tailored to infants' evolving needs, supports healthy growth consistent with WHO standards, for the first year of life.


Subject(s)
Child Development , Infant Formula/adverse effects , Overweight/etiology , Pediatric Obesity/etiology , Urban Health , Body Height/ethnology , Body Mass Index , Cohort Studies , Female , Follow-Up Studies , Growth Charts , Head , Humans , Infant, Newborn , Lost to Follow-Up , Male , Nutritive Value , Overweight/epidemiology , Overweight/ethnology , Patient Dropouts/ethnology , Pediatric Obesity/epidemiology , Pediatric Obesity/ethnology , Risk Factors , Switzerland/epidemiology , Urban Health/ethnology , Weight Gain/ethnology , World Health Organization
8.
Pediatr Res ; 81(4): 622-631, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28002391

ABSTRACT

BACKGROUND: Prebiotics and probiotics exert beneficial effects by modulating gut microbiota and immune system. This study evaluates efficacy and safety of an infant formula containing bovine milk-derived oligosaccharides and Bifidobacterium animalis ssp lactis (B. lactis) (CNCM I-3446) on incidence of diarrhea and febrile infections during the first year of life (primary outcome). METHODS: Full-term infants receiving Test or Control (without bovine milk-derived oligosaccharide and B. lactis) formulae were enrolled in a multicenter, randomized, controlled, and double-blind trial with a reference breastfeeding group. . RESULTS: 413 infants were assigned between Test (n = 206) and Control (n = 207) formula. There was no significant difference for diarrhea and febrile infections incidence between groups at 6 (odds ratio (95% confidence interval) = 0.56 (0.26-1.15), P = 0.096) and 12 mo (odds ratio = 0.66 (0.38-1.14), P = 0.119). Test formula was well tolerated, anthropometrics parameters were not significantly different between groups and aligned with WHO growth standards up to 12 mo. Data from test group showed that gut microbiota pattern, fecal IgA and stool pH were brought to be closer to those of breastfed infants. CONCLUSION: An infant formula enriched with bovine milk-derived oligosaccharide and B. lactis supports normal infant growth, is well tolerated and improves intestinal health markers. No differences in diarrhea and febrile infection incidence were found in the population studied.


Subject(s)
Infant Formula/chemistry , Intestines/physiology , Prebiotics , Probiotics/therapeutic use , Animals , Bifidobacterium animalis , Breast Feeding , Cattle , Diarrhea/microbiology , Double-Blind Method , Fever , Gastrointestinal Microbiome , Humans , Hydrogen-Ion Concentration , Immune System , Infant, Newborn , Kaplan-Meier Estimate , Milk/chemistry , Milk, Human/chemistry , Odds Ratio , Oligosaccharides/chemistry , Treatment Outcome
9.
Clin Med Insights Pediatr ; 10: 119-130, 2016.
Article in English | MEDLINE | ID: mdl-28096702

ABSTRACT

The gut microbiota of infants is shaped by both the mode of delivery and the type of feeding. The gut of vaginally and cesarean-delivered infants is colonized at different rates and with different bacterial species, leading to differences in the gut microbial composition, which may persist up to 6 months. In a multicenter, randomized, controlled, double-blind trial conducted in South Africa, we tested the effect of a formula supplemented with a prebiotic (a mixture of bovine milk-derived oligosaccharides [BMOS] generated from whey permeate and containing galactooligosaccharides and milk oligosaccharides such as 3'- and 6'-sialyllactose) and the probiotic Bifidobacterium animalis subsp. lactis (B. lactis) strain CNCM I-3446 on the bifidobacteria levels in the gut of infants born vaginally or via cesarean section in early life. Additionally, the safety of the new formulation was evaluated. A total of 430 healthy, full-term infants born to HIV-positive mothers who had elected to feed their child beginning from birth (≤3 days old) exclusively with formula were randomized into this multicenter trial of four parallel groups. A total of 421 infants who had any study formula intake were included in the full analysis set (FAS). The first two groups consisted of cesarean-delivered infants assigned to the Test formula (n = 92) (a starter infant formula [IF] containing BMOS at a total oligosaccharide concentration of 5.8 ± 1.0 g/100 g of powder formula [8 g/L in the reconstituted formula] + B. lactis [1 × 107 colony-forming units {cfu}/g]) or a Control IF (n = 101); the second two groups consisted of vaginally delivered infants randomized to the same Test (n = 115) or Control (n = 113) formulas from the time of enrollment to 6 months. The primary efficacy outcome was fecal bifidobacteria count at 10 days, and the primary safety outcome was daily weight gain (g/d) between 10 days and 4 months. At 10 days, fecal bifidobacteria counts were significantly higher in the Test formula than in the Control formula group among infants with cesarean birth (median [range] log: 9.41 [6.30-10.94] cfu/g versus 6.30 [6.30-10.51] cfu/g; P = 0.002) but not among those with vaginal birth (median [range] log: 10.06 [5.93-10.77] cfu/g versus 9.85 [6.15-10.79] cfu/g; P = 0.126). The lower bound of the two-sided 95% confidence interval of the difference in the mean daily weight gain between the Test and Control formula groups was more than -3 g/d in both the vaginally and cesarean-delivered infants, indicating that growth in the Test formula-fed infants was not inferior to that of Control formula-fed infants. At 10 days and 4 weeks, the fecal pH of infants fed the Test formula was significantly lower than in those fed the Control formula, irrespective of mode of delivery: for vaginal delivery: 4.93 versus 5.59; P < 0.001 (10 days) and 5.01 versus 5.71; P < 0.001 (4 weeks); for cesarean delivery: 5.14 versus 5.65, P = 0.009 (10 days) and 5.06 versus 5.75, P < 0.001 (4 weeks). At 3 months, this acidification effect only persisted among cesarean-born infants. IF supplemented with the prebiotic BMOS and probiotic B. lactis induced a strong bifidogenic effect in both delivering modes, but more explicitly correcting the low bifidobacteria level found in cesarean-born infants from birth. The supplemented IF lowered the fecal pH and improved the fecal microbiota in both normal and cesarean-delivered infants. The use of bifidobacteria as a probiotic even in infants who are immunologically at risk is safe and well tolerated.

10.
Environ Microbiol ; 18(7): 2185-95, 2016 07.
Article in English | MEDLINE | ID: mdl-26626365

ABSTRACT

Non-digestible milk oligosaccharides were proposed as receptor decoys for pathogens and as nutrients for beneficial gut commensals like bifidobacteria. Bovine milk contains oligosaccharides, some of which are structurally identical or similar to those found in human milk. In a controlled, randomized double-blinded clinical trial we tested the effect of feeding a formula supplemented with a mixture of bovine milk-derived oligosaccharides (BMOS) generated from whey permeate, containing galacto-oligosaccharides and 3'- and 6'-sialyllactose, and the probiotic Bifidobacterium animalis subsp. lactis (B. lactis) strain CNCM I-3446. Breastfed infants served as reference group. Compared with a non-supplemented control formula, the test formula showed a similar tolerability and supported a similar growth in healthy newborns followed for 12 weeks. The control, but not the test group, differed from the breast-fed reference group by a higher faecal pH and a significantly higher diversity of the faecal microbiota. In the test group the probiotic B. lactis increased by 100-fold in the stool and was detected in all supplemented infants. BMOS stimulated a marked shift to a bifidobacterium-dominated faecal microbiota via increases in endogenous bifidobacteria (B. longum, B. breve, B. bifidum, B. pseudocatenulatum).


Subject(s)
Bifidobacterium animalis/metabolism , Gastrointestinal Microbiome , Infant Formula/analysis , Milk/chemistry , Oligosaccharides/metabolism , Synbiotics/analysis , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Bacteria/isolation & purification , Bifidobacterium animalis/genetics , Bifidobacterium animalis/growth & development , Bifidobacterium animalis/isolation & purification , Cattle , Feces/microbiology , Female , Food Additives/analysis , Food Additives/metabolism , Humans , Infant , Infant, Newborn , Male , Milk/metabolism , Oligosaccharides/analysis
11.
Article in English | MEDLINE | ID: mdl-27057326

ABSTRACT

BACKGROUND: The safety of an infant formula containing a new mixture of the prebiotics galacto-oligosaccharides (GOS) and fructo-oligosaccharide (FOS) and the probiotic Lactobacillus reuteri needs to be evaluated. METHODS: Healthy term infants in Singapore were randomly assigned (using computer-generated allocation sequences) to receive exclusively an experimental infant formula containing L. reuteri, GOS (5.50 g/L), and FOS (0.36 g/L) or a control formula containing only L. reuteri from enrollment (7-14 days of age) to 4 months of age. The primary objective of this trial was to demonstrate that weight change between birth and 4 months of age in infants fed the experimental formula was not inferior to World Health Organization (WHO) Child Growth standards. The non-inferiority margin was -0.5 standard deviations (SD). The secondary objectives were to compare changes in anthropometric measurements (weight, length, body mass index, and head circumference), digestive tolerance, stool bacterial counts, urinary D- and L- lactate concentrations, and adverse events in the two formula groups. RESULTS: The intention-to-treat (ITT) population included all randomized infants stratified by gender, (experimental group, N = 68 and control group, N = 72). The per-protocol (PP) population included 61 infants in the experimental and 62 infants in the control groups. The change in weight-for-age z-score between birth and 4 months was +0.93 (95% confidence interval [CI]: +0.63 to +1.23) SD in the experimental group and +0.92 (95% CI: +0.62 to +1.22) SD in the control group in the PP population, indicating non-inferior weight gain in both formulas groups compared with WHO standards. The ITT population had similar results. Liquid stools occurred more frequently in the experimental compared with the control group and median bifidobacteria, lactobacilli, and enterococci counts were higher in the experimental group (p < 0.05). Other secondary outcomes were not significantly different between groups. CONCLUSIONS: Infant formula containing L. reuteri + GOS/FOS supports normal growth and is safe. TRIAL REGISTRATION: ClinicalTrial.gov: NCT01010113.

12.
BMC Pediatr ; 14: 306, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25527244

ABSTRACT

BACKGROUND: A limited number of nondigestible oligosaccharides are available for use in infant formula. This study evaluated growth and safety in infants fed formula supplemented with a mixture of bovine milk-derived oligosaccharides (BMOS). This mixture, which was generated from whey permeate, contains galactooligosaccharides and other oligosaccharides from bovine milk, such as 3'- and 6'-sialyllactose. We hypothesized that growth in infants fed BMOS-supplemented formula would be noninferior to that in infants fed standard formula. METHODS: Healthy term infants ≤14 days old were randomly assigned to standard formula (control; n = 84); standard formula with BMOS (IF-BMOS; n = 99); or standard formula with BMOS and probiotics (Bifidobacterium longum, Lactobacillus rhamnosus) (IF-BMOS + Pro; n = 98). A breastfed reference group was also enrolled (n = 30). The primary outcome was mean weight gain/day from enrollment to age 4 months (noninferiority margin: -3.0 g/day). RESULTS: 189 (67.3%) formula-fed infants were included in the primary analysis. Mean differences in weight gain between the control and IF-BMOS and IF-BMOS + Pro groups were <1 g/day, with 97.5% confidence intervals above -3.0 g/day, indicating noninferior weight gain in the BMOS formula groups. Compared with control, infants in the BMOS groups had more frequent (p < 0.0001) and less hard (p = 0.0003) stools. No significant differences were observed between the control and BMOS groups in caregivers' reports of flatulence, vomiting, spitting up, crying, fussing, and colic. When based on clinical evaluation by the investigator, the incidence of colic was higher (p = 0.01) in IF-BMOS than in control; the incidence of investigator-diagnosed colic was not significantly different in control and IF-BMOS + Pro (p = 0.15). Stool bifidobacteria and lactobacilli counts were higher with IF-BMOS + Pro compared with control (p < 0.05), whereas Clostridia counts were lower (p < 0.05) in both BMOS groups compared with control. CONCLUSIONS: Infant formula containing BMOS either with or without probiotics provides adequate nutrition for normal growth in healthy term infants. Further studies are needed to fully explore the digestive tolerance of BMOS formula. TRIAL REGISTRATION: ClinicalTrials.gov NCT01886898 . Registered 24 June 2013.


Subject(s)
Child Development , Growth , Infant Formula , Milk , Oligosaccharides/administration & dosage , Animals , Anthropometry , Cattle , Double-Blind Method , Feces/microbiology , Female , Food, Fortified , Gastrointestinal Transit , Humans , Infant , Infant Formula/chemistry , Infant, Newborn , Male , Probiotics/administration & dosage , Weight Gain
13.
Clin Med Insights Pediatr ; 8: 51-60, 2014.
Article in English | MEDLINE | ID: mdl-25452707

ABSTRACT

OBJECTIVE: This multicenter non-inferiority study evaluated the safety of infant formulas enriched with bovine milk fat globule membrane (MFGM) fractions. METHODS: Healthy, full-term infants (n = 119) age ≤14 days were randomized to standard infant formula (control), standard formula enriched with a lipid-rich MFGM fraction (MFGM-L), or standard formula enriched with a protein-rich MFGM fraction (MFGM-P). Primary outcome was mean weight gain per day from enrollment to age 4 months (non-inferiority margin: -3.0 g/day). Secondary (length, head circumference, tolerability, morbidity, adverse events) and exploratory (phospholipids, metabolic markers, immune markers) outcomes were also evaluated. RESULTS: Weight gain was non-inferior in the MFGM-L and MFGM-P groups compared with the control group. Among secondary and exploratory outcomes, few between-group differences were observed. Formula tolerance rates were high (>94%) in all groups. Adverse event and morbidity rates were similar across groups except for a higher rate of eczema in the MFGM-P group (13.9% vs control [3.5%], MFGM-L [1.4%]). CONCLUSION: Both MFGM-enriched formulas met the primary safety endpoint of non-inferiority in weight gain and were generally well tolerated, although a higher rate of eczema was observed in the MFGM-P group.

14.
Pediatr Infect Dis J ; 32(8): 810-4, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23429555

ABSTRACT

BACKGROUND: Several studies have suggested that probiotics (proB) and/or prebiotics (preB) could reduce the burden of infection in infants and toddlers. We aimed to determine whether follow-up formula supplemented with proB and preB could reduce the risk of acute otitis media (AOM). METHODS: In this double-blind, placebo-controlled trial from November 2007 to April 2009, 37 pediatricians in France enrolled children 7 to 13 months of age with high risk of AOM who were randomly assigned to receive follow-up formula supplemented with proB (Streptococcus thermophilus NCC 2496, Streptococcus salivarius DSM 13084, Lactobacillus rhamnosus LPR CGMCC 1.3724) and preB (Raftilose/Raftiline) or follow-up formula alone (placebo). During 12 months, the 2 groups were compared for number of AOM episodes diagnosed (primary outcome) and secondary outcomes by the Poisson model (incidence rate ratio [IRR]) or logistic regression (odds ratio; and 95% confidence interval [95% CI]) after adjustment on covariates of interest. RESULTS: We enrolled 224 children (112 in each group). All children were vaccinated (4 doses) with the 7-valent pneumococcal conjugate vaccine; demographic characteristics were similar in the 2 groups. In total, 486 AOM episodes were reported, 249 and 237 in the treatment and control groups, respectively. The treatment and control groups did not differ in incidence of AOM (IRR 1.0, 95% CI: 0.8-1.2), lower respiratory tract infections (IRR 0.9, 0.7-1.2) or number of antibiotic treatment courses (IRR = 1.0, 95% CI: 0.8-1.2). Treatment was not associated with recurrent AOM (odds ratio 1.0, 95% CI: 0.5-1.7). With regard to gastrointestinal disorders, both formulas were well tolerated. CONCLUSION: The proB and preB included in follow-up formula given to children at 7 to 13 months of age did not reduce the risk of AOM, recurrent AOM, antibiotic use or lower respiratory tract infections at 1 year.


Subject(s)
Otitis Media/drug therapy , Prebiotics , Probiotics/therapeutic use , Acute Disease , Bacteria/classification , Bacteria/isolation & purification , Bacterial Infections/epidemiology , Bacterial Infections/microbiology , Carrier State/epidemiology , Carrier State/microbiology , Chi-Square Distribution , Double-Blind Method , Humans , Incidence , Infant , Nasopharynx/microbiology , Otitis Media/microbiology , Otitis Media/prevention & control , Pneumococcal Vaccines/administration & dosage , Treatment Outcome , Vaccines, Conjugate/administration & dosage
15.
Clin Diagn Lab Immunol ; 11(2): 266-71, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15013974

ABSTRACT

We have previously demonstrated that Lactobacillus paracasei NCC2461 may help to prevent cow's milk allergy in mice by inducing oral tolerance to beta-lactoglobulin (BLG). To investigate the mechanisms involved in this beneficial effect, we examined the possibility that L. paracasei induces tolerance by hydrolyzing BLG-derived peptides and liberating peptides that stimulate interleukin-10 (IL-10) production. L. paracasei peptidases have been shown to hydrolyze tryptic-chymotryptic peptides from BLG, releasing numerous small peptides with immunomodulating properties. We have now shown that acidic tryptic-chymotryptic peptides stimulate splenocyte proliferation and gamma interferon (IFN-gamma) production in vitro. Hydrolysis of these peptides with L. paracasei peptidases repressed the lymphocyte stimulation, up-regulated IL-10 production, and down-regulated IFN-gamma and IL-4 secretion. L. paracasei NCC2461 may therefore induce oral tolerance to BLG in vivo by degrading acidic peptides and releasing immunomodulatory peptides stimulating regulatory T cells, which function as major immunosuppressive agents by secreting IL-10.


Subject(s)
Immune Tolerance/immunology , Interleukin-10/metabolism , Lactobacillus/enzymology , Lactobacillus/immunology , Lactoglobulins/immunology , Lactoglobulins/metabolism , Animals , Bacterial Proteins/metabolism , Cell Division/immunology , Female , Hydrolysis , Interferon-gamma/metabolism , Lymphocytes/cytology , Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Peptide Hydrolases/metabolism , Spleen/immunology
16.
Ann N Y Acad Sci ; 1029: 374-8, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15681785

ABSTRACT

Ibuprofen and antibiotics are commonly prescribed during early childhood. When given to mice at the time at which oral tolerance is induced, both treatments affect either the induction or the maintenance of oral tolerance. These results suggest that the coadministration of these and similarly acting drugs should be considered cautiously for infants at risk of allergy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Ibuprofen/pharmacology , Immune Tolerance/immunology , Animals , Child, Preschool , Dinoprostone/metabolism , Feces/microbiology , Humans , Immune Tolerance/drug effects , Immunoglobulin E/blood , Mice , Mice, Inbred BALB C
17.
Clin Diagn Lab Immunol ; 10(5): 787-92, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12965905

ABSTRACT

In this study, the effect of Lactobacillus paracasei (NCC 2461), Lactobacillus johnsonii (NCC 533) and Bifidobacterium lactis Bb12 (NCC 362) on the induction and maintenance of oral tolerance to bovine beta-lactoglobulin (BLG) was investigated in mice. Germfree mice were monocolonized with one of the three strains before oral administration of whey protein to induce tolerance. Mice were then injected with BLG and sacrificed 28 or 50 days after whey protein feeding for humoral and cellular response measurement. Conventional and germfree mice were used as controls. Both humoral and cellular responses were better suppressed in conventional mice than in germfree and monoassociated mice throughout the experiment and better suppressed in L. paracasei-associated mice than in mice colonized with B. lactis or L. johnsonii. The latter two mono-associations suppressed humoral responses only partially and cellular responses not at all. This study provides evidence that probiotics modulate the oral tolerance response to BLG in mice. The mono-colonization effect is strain-dependant, the best result having been obtained with L. paracasei.


Subject(s)
Germ-Free Life , Immune Tolerance , Lactoglobulins/immunology , Probiotics , Administration, Oral , Animals , Bifidobacterium/immunology , Feces/microbiology , Intestines/immunology , Intestines/microbiology , Lactobacillus/immunology , Mice , Milk Proteins/administration & dosage , Milk Proteins/immunology , Whey Proteins
18.
Int J Pharm ; 242(1-2): 15-24, 2002 Aug 21.
Article in English | MEDLINE | ID: mdl-12176221

ABSTRACT

Mucosal administration of antigen is known to be appropriate for vaccine purposes as well as tolerance induction. Biodegradable poly(DL-lactide-co-glycolide) (PLGA) microparticles were used to deliver both antibacterial phosphorylcholine (PC) and dietary antigen beta lactoglobulin (BLG) by mucosal route. In a first study, the protective immunity elicited by intragastric vaccination with PC encapsulated in microparticles was evaluated in a mouse model against intestinal infection by Salmonella typhimurium and pulmonary infection by Streptococcus pneumoniae. A significant rise in anti-PC immunoglobulin A (IgA) titers, as measured by an enzyme-linked immunosorbent assay, was observed in the intestinal secretions after oral immunization with PC-loaded microparticles compared with the titers of mice immunized with free PC-thyr or blank microparticles. This antibody response correlated with a highly significant resistance to oral challenge by S. typhimurium. IgA in pulmonary secretion were not able to protect against S. pneumoniae infection. BALB/c mice were, therefore, immunized intranasally (i.n.). Immunization was followed by a rise in anti-PC IgA and IgG titers in serum and in pulmonary secretions by both free and encapsulated PC-Thyr. The survival rates were 91 and 76% in the two groups of mice, respectively. In a second study and in order to prevent allergy against milk by inducing oral tolerance, one of the major allergenic milk protein, BLG was entrapped into microparticles. Oral administration of microparticles containing BLG reduced significantly (by 10000) the amount of protein necessary to decrease both specific anti BLG IgE and DTH response. These studies demonstrate the ability of microparticles to induce both mucosal immunity and oral tolerance.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Antigens/administration & dosage , Food Hypersensitivity/immunology , Lactoglobulins/administration & dosage , Lactoglobulins/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/analysis , Antibodies, Bacterial/biosynthesis , Colony Count, Microbial , Excipients , Female , Immunoglobulins/analysis , Immunoglobulins/biosynthesis , Lactic Acid , Lactoglobulins/adverse effects , Mice , Mice, Inbred BALB C , Microspheres , Particle Size , Pneumococcal Infections/immunology , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers , Salmonella Infections/immunology , Salmonella Infections/prevention & control , Salmonella typhimurium/immunology , Streptococcus pneumoniae/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...