Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Nutrients ; 14(5)2022 Feb 28.
Article in English | MEDLINE | ID: mdl-35267989

ABSTRACT

BACKGROUND: Human milk oligosaccharides are complex, non-digestible carbohydrates that directly interact with intestinal epithelial cells to alter barrier function and host inflammation. Oligosaccharide composition varies widely between individual mothers, but it is unclear if this inter-individual variation has any impact on intestinal epithelial barrier function and gut inflammation. METHODS: Human milk oligosaccharides were extracted from the mature human milk of four individual donors. Using an in vitro model of intestinal injury, the effects of the oligosaccharides on the intestinal epithelial barrier and select innate and adaptive immune functions were assessed. RESULTS: Individual oligosaccharide compositions shared comparable effects on increasing transepithelial electrical resistance and reducing the macromolecular permeability of polarized (Caco-2Bbe1) monolayers but exerted distinct effects on the localization of the intercellular tight junction protein zona occludins-1 in response to injury induced by a human enteric bacterial pathogen Escherichia coli, serotype O157:H7. Immunoblots showed the differential effects of oligosaccharide compositions in reducing host chemokine interleukin 8 expression and inhibiting of p38 MAP kinase activation. CONCLUSIONS: These results provide evidence of both shared and distinct effects on the host intestinal epithelial function that are attributable to inter-individual differences in the composition of human milk oligosaccharides.


Subject(s)
Intestinal Mucosa , Milk, Human , Humans , Inflammation/metabolism , Intestinal Mucosa/metabolism , Oligosaccharides/pharmacology , Pilot Projects
2.
Pediatr Allergy Immunol ; 31(5): 544-553, 2020 07.
Article in English | MEDLINE | ID: mdl-32150651

ABSTRACT

BACKGROUND: Perinatal childhood exposures, including probiotic supplementation, may affect epigenetic modifications and impact on immune maturation and allergy development. The aim of this study was to assess the effects of pre- and postnatal Lactobacillus reuteri supplementation on DNA methylation in relation to immune maturation and allergy development. METHODS: DNA methylation patterns were investigated for allergy-related T helper subsets using a locus-specific method and at a genome-wide scale using the Illumina 450K array. From a randomised, double-blind, placebo-controlled allergy prevention trial with pre- and postnatal probiotic supplementation, CD4+ T helper cells were obtained at birth (from cord blood), and 12 and 24 months of age (total (placebo/probiotics); locus-specific method: CB = 32 (17/15), 12 months = 24 (9/15), 24 months = 35 (15/20); Illumina: CB = 19 (10/9), 12 months = 10 (6/4), 24 months = 19(11/8)). RESULTS: Comparing probiotics to placebo, the greatest genome-wide differential DNA methylation was observed at birth, where the majority of sites were hypomethylated, indicating transcriptional accessibility in the probiotic group. Bioinformatic analyses, including network analyses, revealed a module containing 91 genes, enriched for immune-related pathways such as chemotaxis, PI3K-Akt, MAPK and TGF-ß signalling. A majority of the module genes were associated with atopic manifestations (OR = 1.43, P = 2.4 × 10-6 ), and a classifier built on this model could predict allergy development (AUC = 0.78, P = 3.0 × 10e-3 ). Pathways such as IFN-γ signalling and T-cell activation were more hypermethylated at birth compared with later in life in both intervention groups over time, in line with DNA methylation patterns in the IFNG locus obtained by the locus-specific methodology. CONCLUSION: Maternal L. reuteri supplementation during pregnancy alters DNA methylation patterns in CD4+ T cells towards enhanced immune activation at birth, which may affect immune maturation and allergy development.


Subject(s)
Hypersensitivity , Limosilactobacillus reuteri , Probiotics , Child , Child, Preschool , DNA Methylation , Double-Blind Method , Female , Humans , Infant , Infant, Newborn , Phosphatidylinositol 3-Kinases , Pregnancy , T-Lymphocytes, Helper-Inducer
3.
Pediatr Allergy Immunol ; 31(3): 250-257, 2020 04.
Article in English | MEDLINE | ID: mdl-31736150

ABSTRACT

BACKGROUND: Early colonization with a diverse microbiota seems to play a crucial role for appropriate immune maturation during childhood. Breastmilk microbiota is one important source of microbes for the infant, transferred together with maternal IgA antibodies. We previously observed that allergy development during childhood was associated with aberrant IgA responses to the gut microbiota already at 1 month of age, when the IgA antibodies are predominantly maternally derived in breastfed infants. OBJECTIVE: To determine the microbial composition and IgA-coated bacteria in breastmilk in relation to allergy development in children participating in an intervention trial with pre- and post-natal Lactobacillus reuteri supplementation. METHODS: A combination of flow cytometric cell sorting and 16S rRNA gene sequencing was used to characterize the bacterial recognition patterns by IgA in breastmilk samples collected one month post-partum from 40 mothers whose children did or did not develop allergic and asthmatic symptoms during the first 7 years of age. RESULTS: The milk fed to children developing allergic manifestations had significantly lower bacterial richness, when compared to the milk given to children that remained healthy. Probiotic treatment influenced the breastmilk microbiota composition. However, the proportions of IgA-coated bacteria, the total bacterial load and the patterns of IgA-coating were similar in breastmilk between mothers of healthy children and those developing allergies. CONCLUSION: Consumption of breastmilk with a reduced microbial richness in the first month of life may play an important role in allergy development during childhood.


Subject(s)
Hypersensitivity/epidemiology , Milk, Human/immunology , Milk, Human/microbiology , Asthma/epidemiology , Breast Feeding , Child , Child, Preschool , Female , Flow Cytometry , Food Hypersensitivity/epidemiology , Gastrointestinal Microbiome , Humans , Hypersensitivity/immunology , Hypersensitivity/microbiology , Immunoglobulin A/immunology , Infant , Infant, Newborn , Male , Microbiota/genetics , Probiotics/therapeutic use , RNA, Ribosomal, 16S
4.
Mol Nutr Food Res ; 63(3): e1800658, 2019 02.
Article in English | MEDLINE | ID: mdl-30407734

ABSTRACT

SCOPE: Necrotizing enterocolitis (NEC) is a leading cause of morbidity and death in preterm infants, occurring more often in formula-fed than breastfed infants. Studies in both rats and humans show that human milk oligosaccharides (HMOs) lower the incidence of NEC, but the mechanism underlying such protection is currently unclear. METHODS AND RESULTS: By extracting HMOs from pooled human breastmilk, the impact of HMOs on the intestinal mucin levels in a murine model of NEC are investigated. To confirm the results, the findings are validated by exposing human intestinal epithelial cells and intestinal organoids to HMOs and evaluated for mucin expression. HMO-gavage to pups increases Muc2 levels and decreases intestinal permeability to macromolecular dextran. HMO-treated cells have increased Muc2 expression, decreased bacterial attachment and dextran permeability during challenge by enteric pathogens. To identify the mediators involved in HMO induction of mucins, it is demonstrated that HMOs directly induce the expression of chaperone proteins including protein disulfide isomerase (PDI). Suppression of PDI activity removes the protective effects of HMOs on barrier function in vitro as well as NEC protection in vivo. CONCLUSIONS: Taken together, the results provide insights to the possible mechanisms by which HMOs protect the neonatal intestine through upregulation of mucins.


Subject(s)
Enterocolitis, Necrotizing/prevention & control , Milk, Human/chemistry , Mucin-2/genetics , Oligosaccharides/pharmacology , Animals , Animals, Newborn , Caco-2 Cells , Endoplasmic Reticulum Stress/drug effects , Enterocolitis, Necrotizing/metabolism , Goblet Cells/drug effects , Humans , Intestinal Mucosa/drug effects , Mice , Mice, Inbred C57BL , Mucin-2/analysis , Protein Disulfide-Isomerases/physiology
5.
Nutrients ; 10(10)2018 Oct 20.
Article in English | MEDLINE | ID: mdl-30347801

ABSTRACT

Difference in human milk oligosaccharides (HMO) composition in breast milk may be one explanation why some preterm infants develop necrotizing enterocolitis (NEC) despite being fed exclusively with breast milk. The aim of this study was to measure the concentration of 15 dominant HMOs in breast milk during the neonatal period and investigate how their levels correlated to NEC, sepsis, and growth in extremely low birth weight (ELBW; <1000 g) infants who were exclusively fed with breast milk. Milk was collected from 91 mothers to 106 infants at 14 and 28 days and at postmenstrual week 36. The HMOs were analysed with high-performance anion-exchange chromatography with pulsed amperometric detection. The HMOs diversity and the levels of Lacto-N-difucohexaose I were lower in samples from mothers to NEC cases, as compared to non-NEC cases at all sampling time points. Lacto-N-difucohexaose I is only produced by secretor and Lewis positive mothers. There were also significant but inconsistent associations between 3'-sialyllactose and 6'-sialyllactose and culture-proven sepsis and significant, but weak correlations between several HMOs and growth rate. Our results suggest that the variation in HMO composition in breast milk may be an important factor explaining why exclusively breast milk fed ELBW infants develop NEC.


Subject(s)
Enterocolitis, Necrotizing/etiology , Infant, Extremely Low Birth Weight , Milk, Human/chemistry , Oligosaccharides/adverse effects , Oligosaccharides/classification , Humans , Infant, Newborn , Oligosaccharides/chemistry
7.
Nestle Nutr Inst Workshop Ser ; 88: 107-115, 2017.
Article in English | MEDLINE | ID: mdl-28346927

ABSTRACT

The complex and diverse intestinal microbiome is recognized as important in promoting human health. An altered gut microflora, referred to as dysbiosis, is increasingly recognized as having an etiologic role in a variety of conditions, including functional gastrointestinal disorders: colic in infants and irritable bowel syndrome in older children. Probiotics are defined as live microorganisms that, if ingested in sufficient amounts, restore microbial homeostasis and have a benefit on health. Randomized controlled trials indicate that probiotics can be effective in a variety of intestinal conditions, including colic and irritable bowel syndrome. Probiotics may promote gut microbial diversity, but timing of the intervention appears crucial. Strain-specific effects on colonization resistance, epithelial barrier integrity, modulation of signal transduction, impacts on innate and adaptive immune responses, and effects on visceral hyperalgesia likely explain the observed variability in various probiotic strains. In the future, probiotics are likely to be chosen for use in a defined clinical setting based on underlying mechanism(s) of action. The precise component of the probiotic agent mediating observed effects is the subject of current research. Unresolved issues relate to optimal dosages, timing of ingestion, single versus combination formulations, maintenance of viability in storage, and the merits of employing probiotic-derived products.


Subject(s)
Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome/physiology , Animals , Bifidobacterium , Child , Colic/microbiology , Colic/therapy , Dysbiosis , Feces/microbiology , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/immunology , Humans , Infant , Infant, Newborn , Irritable Bowel Syndrome/microbiology , Lactobacillus , Probiotics/therapeutic use
8.
J Allergy Clin Immunol ; 139(3): 1017-1025.e14, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27531072

ABSTRACT

BACKGROUND: Although a reduced gut microbiota diversity and low mucosal total IgA levels in infancy have been associated with allergy development, IgA responses to the gut microbiota have not yet been studied. OBJECTIVE: We sought to determine the proportions of IgA coating together with the characterization of the dominant bacteria, bound to IgA or not, in infant stool samples in relation to allergy development. METHODS: A combination of flow cytometric cell sorting and deep sequencing of the 16S rDNA gene was used to characterize the bacterial recognition patterns by IgA in stool samples collected at 1 and 12 months of age from children staying healthy or having allergic symptoms up to 7 years of age. RESULTS: The children with allergic manifestations, particularly asthma, during childhood had a lower proportion of IgA bound to fecal bacteria at 12 months of age compared with healthy children. These alterations cannot be attributed to differences in IgA levels or bacterial load between the 2 groups. Moreover, the bacterial targets of early IgA responses (including coating of the Bacteroides genus), as well as IgA recognition patterns, differed between healthy children and children with allergic manifestations. Altered IgA recognition patterns in children with allergy were observed already at 1 month of age, when the IgA antibodies are predominantly maternally derived in breast-fed children. CONCLUSION: An aberrant IgA responsiveness to the gut microbiota during infancy precedes asthma and allergy development, possibly indicating an impaired mucosal barrier function in allergic children.


Subject(s)
Feces/microbiology , Gastrointestinal Microbiome , Hypersensitivity/immunology , Hypersensitivity/microbiology , Immunoglobulin A/immunology , Bacteria/isolation & purification , Bacterial Load , Child , Child, Preschool , Female , Humans , Infant , Male
9.
Adv Nutr ; 7(5): 928-37, 2016 09.
Article in English | MEDLINE | ID: mdl-27633108

ABSTRACT

Necrotizing enterocolitis (NEC) is a devastating intestinal disease in preterm infants characterized by barrier disruption, intestinal microbial dysbiosis, and persistent inflammation of the colon, which results in high mortality rates. Current strategies used to manage this disease are not sufficient, although the use of human breast milk reduces the risk of NEC. Mother's milk is regarded as a fundamental nutritional source for neonates, but pasteurization of donor breast milk affects the composition of bioactive compounds. Current research is evaluating the benefits and potential pitfalls of adding probiotics and prebiotics to pasteurized milk so as to improve the functionality of the milk and thereby reduce the burden of illness caused by NEC. Probiotics (live micro-organisms that confer health to the host) and prebiotics (nondigestible oligosaccharides that stimulate the growth of healthy bacteria) are functional foods known to mediate immune responses and modulate microbial populations in the gut. Clinical research shows strain- and compound-specific responses when probiotics or prebiotics are administered in conjunction with donor breast milk for the prevention of NEC. Despite ongoing controversy surrounding optimal treatment strategies, randomized controlled studies are now investigating the use of synbiotics to reduce the incidence and severity of NEC. Synbiotics, a combination of probiotics and prebiotics, have been proposed to enhance beneficial health effects in the intestinal tract more than either agent administered alone. This review considers the implications of using probiotic-, prebiotic-, and synbiotic-supplemented breast milk as a strategy to prevent NEC and issues that could be encountered with the preparations.


Subject(s)
Enterocolitis, Necrotizing/prevention & control , Milk, Human , Prebiotics , Probiotics , Synbiotics , Bacteria , Humans , Milk, Human/chemistry , Milk, Human/microbiology , Neonatology , Oligosaccharides
11.
J Allergy Clin Immunol ; 136(5): 1378-86.e1-5, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26014812

ABSTRACT

BACKGROUND: Although immune responses directed against antigens from the intestinal microbiota are observed in certain diseases, the normal human adaptive immune response to intestinal microbiota is poorly defined. OBJECTIVE: Our goal was to assess the adaptive immune response to the intestinal microbiota present in 143 healthy adults and compare this response with the response observed in 52 children and their mothers at risk of having allergic disease. METHODS: Human serum was collected from adults and children followed from birth to 7 years of age, and the serum IgG response to a panel of intestinal microbiota antigens was assessed by using a novel protein microarray. RESULTS: Nearly every subject tested, regardless of health status, had serum IgG that recognized a common set of antigens. Seroreactivity to the panel of antigens was significantly lower in atopic adults. Healthy infants expressed the highest level of IgG seroreactivity to intestinal microbiota antigens. This adaptive response developed between 6 and 12 months of age and peaked around 2 years of age. Low IgG responses to certain clusters of microbiota antigens during infancy were associated with allergy development during childhood. CONCLUSIONS: There is an observed perturbation of the adaptive response to antigens from the microbiota in allergic subjects. These perturbations are observable even in childhood, suggesting that optimal stimulation of the adaptive immune system by the microbiota might be needed to prevent certain immune-mediated diseases.


Subject(s)
Antigens, Bacterial/immunology , Gastrointestinal Microbiome/immunology , Hypersensitivity/immunology , Intestines/immunology , Adaptive Immunity , Adult , Child , Child, Preschool , Female , Humans , Immunoglobulin G/blood , Infant , Infant, Newborn , Intestines/microbiology , Male , Microarray Analysis
12.
Pediatr Res ; 77(1-2): 214-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25310761

ABSTRACT

Limited microbial exposure is suggested to underlie the increase of allergic diseases in affluent countries, and bacterial diversity seems to be more important than specific bacteria taxa. Prospective studies indicate that the gut microbiota composition during the first months of life influences allergy development, and support the theory that factors influencing the early maturation of the immune system might be important for subsequent allergic disease. However, recent research indicates that microbial exposure during pregnancy may be even more important for the preventative effects against allergic disease. This review gives a background of the epidemiology, immunology, and microbiology literature in this field. It focuses on possible underlying mechanisms such as immune-regulated epigenetic imprinting and bacterial translocation during pregnancy, potentially providing the offspring with a pioneer microbiome. We suggest that a possible reason for the initial exposure of bacterial molecular patterns to the fetus in utero is to prime the immune system and/or the epithelium to respond appropriately to pathogens and commensals after birth.


Subject(s)
Epigenesis, Genetic/immunology , Gastrointestinal Tract/microbiology , Hypersensitivity/epidemiology , Hypersensitivity/immunology , Hypersensitivity/microbiology , Microbiota , Prenatal Exposure Delayed Effects/microbiology , Female , Humans , Models, Biological , Pregnancy
13.
Clin Transl Allergy ; 4: 21, 2014.
Article in English | MEDLINE | ID: mdl-25002964

ABSTRACT

BACKGROUND: Mice models indicate that intact Toll like receptor (TLR) signaling may be essential for the allergy protective effects of diverse bacterial exposure observed in clinical trials and epidemiological studies. Probiotic supplementation with Lactobacillus reuteri from pregnancy week 36 and to the infant through the first year of life decreased the prevalence of IgE-associated eczema at two years (ClinicalTrials.gov NCT01285830). The effect of this supplementation on innate immune responses to bacterial products and the expression of associated TLRs were explored. METHODS: Blood mononuclear cells were collected at birth, 6, 12 and 24 months from 61 infants and cultured with TLR2, 4 and 9 ligands. Cytokine and chemokine secretion was determined as well as TLR2, 4 and 9 mRNA expression. RESULTS: Probiotic supplementation was associated with decreased LTA (lipoteichoic acid) induced CCL4, CXCL8, IL-1ß and IL-6 responses at 12 months and decreased CCL4 and IL-1ß secretion at 24 months. TLR2 mRNA expression was not affected by probiotic treatment. CONCLUSIONS: Decreased responses to TLR2, the main receptor for LTA from Gram positive bacteria, in probiotic treated children seem to be dependent on factors downstream of TLR mRNA expression.

15.
Gut ; 63(4): 559-66, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23926244

ABSTRACT

OBJECTIVE: The early intestinal microbiota exerts important stimuli for immune development, and a reduced microbial exposure as well as caesarean section (CS) has been associated with the development of allergic disease. Here we address how microbiota development in infants is affected by mode of delivery, and relate differences in colonisation patterns to the maturation of a balanced Th1/Th2 immune response. DESIGN: The postnatal intestinal colonisation pattern was investigated in 24 infants, born vaginally (15) or by CS (nine). The intestinal microbiota were characterised using pyrosequencing of 16S rRNA genes at 1 week and 1, 3, 6, 12 and 24 months after birth. Venous blood levels of Th1- and Th2-associated chemokines were measured at 6, 12 and 24 months. RESULTS: Infants born through CS had lower total microbiota diversity during the first 2 years of life. CS delivered infants also had a lower abundance and diversity of the Bacteroidetes phylum and were less often colonised with the Bacteroidetes phylum. Infants born through CS had significantly lower levels of the Th1-associated chemokines CXCL10 and CXCL11 in blood. CONCLUSIONS: CS was associated with a lower total microbial diversity, delayed colonisation of the Bacteroidetes phylum and reduced Th1 responses during the first 2 years of life.


Subject(s)
Bacteroidetes/physiology , Cesarean Section/adverse effects , Chemokine CXCL10/blood , Chemokine CXCL11/blood , Intestines/microbiology , Microbiota/physiology , Th1 Cells/physiology , Age Factors , Bacteroidetes/genetics , Chemokine CXCL10/physiology , Chemokine CXCL11/physiology , DNA, Bacterial/genetics , Feces/microbiology , Female , Humans , Immunity, Cellular/physiology , Infant , Infant, Newborn , Male , Microbiota/genetics , RNA, Ribosomal, 16S/genetics
17.
Pediatr Allergy Immunol ; 24(6): 556-61, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23902407

ABSTRACT

BACKGROUND: Supplementation with the probiotic Lactobacillus reuteri reduced the incidence of IgE-associated allergic disease in infancy. This treatment might therefore also reduce the risk of asthma and allergic rhinoconjunctivitis in school age. OBJECTIVE: To evaluate whether perinatal and infant supplementation with L. reuteri reduced the prevalence of respiratory allergic disease in school age and to explore whether this supplementation was associated with any long-term side effects. METHODS: A randomized, placebo-controlled trial with oral supplementation with L. reuteri ATCC 55730 (1 × 10(8) CFU) during the last month of gestation and through the first year of life comprising 232 families with allergic disease, of whom 184 completed a 7-yr follow-up. The primary outcomes at 7 yr of age were allergic disease and skin prick test reactivity (ClinicalTrials.gov ID NCT01285830). RESULTS: The prevalence of asthma (15% in the probiotic vs. 16% in placebo group), allergic rhinoconjunctivitis (27% vs. 20%), eczema (21% vs. 19%) and skin prick test reactivity (29% vs. 26%) was similar in the probiotic and placebo group. Growth indices and gastrointestinal symptoms were similar in the two groups. No severe adverse events were reported. CONCLUSION: The effect of L. reuteri on sensitization and IgE-associated eczema in infancy did not lead to a lower prevalence of respiratory allergic disease in school age. Thus, the effect of L. reuteri on the immune system seems to be transient. Administration of L. reuteri during the last weeks of gestation and in infancy was not associated with any long-term side effects.


Subject(s)
Limosilactobacillus reuteri/immunology , Population , Probiotics/administration & dosage , Respiratory Hypersensitivity/epidemiology , Respiratory Hypersensitivity/therapy , Child , Female , Follow-Up Studies , Humans , Infant, Newborn , Male , Prevalence , Probiotics/adverse effects , Time Factors
19.
J Allergy Clin Immunol ; 129(2): 434-40, 440.e1-2, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22153774

ABSTRACT

BACKGROUND: It is debated whether a low total diversity of the gut microbiota in early childhood is more important than an altered prevalence of particular bacterial species for the increasing incidence of allergic disease. The advent of powerful, cultivation-free molecular methods makes it possible to characterize the total microbiome down to the genus level in large cohorts. OBJECTIVE: We sought to assess microbial diversity and characterize the dominant bacteria in stool during the first year of life in relation to atopic eczema development. METHODS: Microbial diversity and composition were analyzed with barcoded 16S rDNA 454-pyrosequencing in stool samples at 1 week, 1 month, and 12 months of age in 20 infants with IgE-associated eczema and 20 infants without any allergic manifestation until 2 years of age (ClinicalTrials.gov ID NCT01285830). RESULTS: Infants with IgE-associated eczema had a lower diversity of the total microbiota at 1 month (P = .004) and a lower diversity of the bacterial phylum Bacteroidetes and the genus Bacteroides at 1 month (P = .02 and P = .01) and the phylum Proteobacteria at 12 months of age (P = .02). The microbiota was less uniform at 1 month than at 12 months of age, with a high interindividual variability. At 12 months, when the microbiota had stabilized, Proteobacteria, comprising gram-negative organisms, were more abundant in infants without allergic manifestation (Empirical Analysis of Digital Gene Expression in R [edgeR] test: P = .008, q = 0.02). CONCLUSION: Low intestinal microbial diversity during the first month of life was associated with subsequent atopic eczema.


Subject(s)
Eczema/microbiology , Hypersensitivity/microbiology , Intestines/microbiology , Allergens/immunology , Bacteria/classification , Bacteria/genetics , Child, Preschool , DNA, Bacterial/analysis , Eczema/diagnosis , Eczema/prevention & control , Feces/microbiology , Female , Food Hypersensitivity/blood , Food Hypersensitivity/diagnosis , Food Hypersensitivity/immunology , Humans , Hypersensitivity/diagnosis , Hypersensitivity/prevention & control , Immunoglobulin E/blood , Infant , Infant, Newborn , Male , Probiotics/therapeutic use , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
20.
J Pediatr Gastroenterol Nutr ; 49(3): 349-54, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19525871

ABSTRACT

OBJECTIVES: This is to identify factors affecting the prevalence of Lactobacillus reuteri in maternal faeces and breast milk and infant faeces after oral supplementation with L reuteri and to assess the influence on microbial ecology, particularly Clostridium difficile and Bifidobacterium colonization. MATERIALS AND METHODS: In this double-blind trial, 232 mothers with a family history of atopic disease were randomized to a daily intake of either L reuteri American-type culture collection (ATCC) 55730 (1 x 10 colony-forming units [CFU]) or placebo for the last 4 weeks of pregnancy. Their babies then continued with the same study product daily from birth until 12 months of age. Bacterial counts and prevalence were assessed in maternal breast milk and faeces and infant faeces, using conventional cultivation methods. RESULTS: The prevalence of L reuteri was higher during the first year of life in the stool samples from infants in the active as compared with the placebo-treated group. The highest prevalence was recorded at 5 to 6 days of age (82% in the treated vs 20% in the placebo group, P < 0.001). Lactobacillus reuteri was isolated from 12% and 2%, respectively, in the colostrum samples (P < 0.05). Breast-feeding seemed to reduce faecal L reuteri counts, although antibiotics did not influence the levels of L reuteri. The administration of L reuteri did not affect bifidobacteria or C difficile colonization. CONCLUSION: Lactobacillus reuteri may be detected in breast milk after oral supplementation to the mother and in almost all infants after oral supplementation during the first year of life, as well as occasionally in many untreated infants.


Subject(s)
Colostrum/microbiology , Dietary Supplements , Feces/microbiology , Limosilactobacillus reuteri/isolation & purification , Milk, Human/microbiology , Probiotics/administration & dosage , Adult , Bifidobacterium/isolation & purification , Clostridioides difficile/isolation & purification , Colony Count, Microbial , Double-Blind Method , Female , Humans , Hypersensitivity , Infant , Pregnancy , Probiotics/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...