Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Mol Nutr Food Res ; 67(15): e2200446, 2023 08.
Article in English | MEDLINE | ID: mdl-37326413

ABSTRACT

SCOPE: Modifying the composition of colostrum by external factors may provide opportunities to improve the infant's health. Here, we evaluated how fish oil and/or probiotics supplementation modify concentrations of colostrum immune mediators and their associations with perinatal clinical factors on mothers with overweight/obesity. METHODS AND RESULTS: Pregnant women were randomized in a double-blind manner into four intervention groups, and the supplements were consumed daily from early pregnancy onwards. Colostrum samples were collected from 187 mothers, and 16 immune mediators were measured using bead-based immunoassays. Interventions modified colostrum composition; the fish oil+probiotics group had higher concentrations of IL-12p70 than probiotics+placebo and higher FMS-like tyrosine kinase 3 ligand (FLT-3L) than fish oil+placebo and probiotics+placebo (one-way analysis of variance, post-hoc Tukey's test). Although the fish oil+probiotics group had higher levels of IFNα2 compared to the fish oil+placebo group, these differences were not statistically significant after correction for multiple testing. Multivariate linear model revealed significant associations between several immune mediators and the perinatal use of medication. CONCLUSION: Fish oil/probiotics intervention exerted a minor effect on concentrations of colostrum immune mediators. However, medication during the perinatal period modulated the immune mediators. These changes in colostrum's composition may contribute to immune system development in the infant.


Subject(s)
Fish Oils , Probiotics , Female , Humans , Pregnancy , Colostrum , Dietary Supplements , Double-Blind Method , Obesity/complications , Overweight/complications , Probiotics/therapeutic use
2.
Br J Nutr ; 128(11): 2181-2192, 2022 12 14.
Article in English | MEDLINE | ID: mdl-35086570

ABSTRACT

Pulses are healthy and sustainable but induce gut symptoms in people with a sensitive gut. Oats, on the contrary, have no fermentable oligo- di-, monosaccharides and polyols compounds and are known for the health effects of their fibres. This 4-day cross-over trial investigated the effects of oat and rice flour ingested with pulses on gut symptoms and exhaled gases (4th day only) in subjects with a sensitive gut or IBS (n 21) and controls (n 21). The sensitive group perceived more symptoms after both meals than controls (P = 0·001, P = 0·001). Frequency, intensity or quality of the symptoms did not differ between meals during the first 3 d in either group. More breath hydrogen was produced after an oat than rice containing meal in both groups (AUC, P = 0·001, P = 0·001). No between-group difference was seen in breath gases. During day 4, both sensitive and control groups perceived more symptoms after the oat flour meal (P = 0·001, P = 0·0104, respectively) as mainly mild flatulence. No difference in moderate or severe symptoms was detected. Increased hydrogen production correlated to a higher amount of perceived flatulence after the oat flour meal in both the sensitive and the control groups (P = 0·042, P = 0·003, respectively). In summary, ingestion of oat flour with pulses increases breath hydrogen levels compared with rice flour, but gastrointestinal symptoms of subjects sensitive to pulses were not explained by breath hydrogen levels. Additionally, consumer mindsets towards pulse consumption and pulse-related gut symptoms were assessed by an online survey, which implied that perceived gut symptoms hinder the use of pulses in sensitive subjects.


Subject(s)
Avena , Gastrointestinal Diseases , Humans , Hydrogen , Flour , Flatulence , Cross-Over Studies , Gases , Breath Tests
3.
Food Chem ; 342: 128219, 2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33077284

ABSTRACT

While the development of oat products often requires altered molecular weight (MW) of ß-glucan, the resulting health implications are currently unclear. This 3-leg crossover trial (n = 14) investigated the effects of the consumption of oat bran with High, Medium and Low MW ß-glucan (average > 1000, 524 and 82 kDa respectively) with 3 consequent meals on oat-derived phenolic compounds in urine (UHPLC-MS/MS), bile acids in feces (UHPLC-QTOF), gastrointestinal conditions (ingestible capsule), and perceived gut well-being. Urine excretion of ferulic acid was higher (p < 0.001, p < 0.001), and the fecal excretion of deoxycholic (p < 0.03, p < 0.02) and chenodeoxycholic (p < 0.06, p < 0.02) acids lower after consumption of Low MW ß-glucan compared with both Medium and High MW ß-glucan. Duodenal pressure was higher after consumption of High MW ß-glucan compared to Medium (p < 0.041) and Low (p < 0.022) MW ß-glucan. The MW of ß-glucan did not affect gut well-being, but the perceptions between females and males differed.


Subject(s)
Bile Acids and Salts/metabolism , Feces/chemistry , Gastrointestinal Tract/drug effects , Urine/chemistry , beta-Glucans/chemistry , beta-Glucans/pharmacology , Cross-Over Studies , Dietary Fiber , Female , Humans , Male , Molecular Weight , Sex Characteristics
4.
Int J Mol Sci ; 21(17)2020 Aug 22.
Article in English | MEDLINE | ID: mdl-32842596

ABSTRACT

Inflammatory bowel diseases (IBD), ulcerative colitis (UC) and Crohn's disease (CD), are chronic debilitating disorders of unknown etiology. Over 200 genetic risk loci are associated with IBD, highlighting a key role for immunological and epithelial barrier functions. Environmental factors account for the growing incidence of IBD, and microbiota are considered as an important contributor. Microbiota dysbiosis can lead to a loss of tolerogenic immune effects and initiate or exacerbate inflammation. We aimed to study colonic mucosal microbiota and the expression of selected host genes in pediatric UC. We used high-throughput 16S rDNA sequencing to profile microbiota in colonic biopsies of pediatric UC patients (n = 26) and non-IBD controls (n = 27). The expression of 13 genes, including five for antimicrobial peptides, in parallel biopsies was assessed with qRT-PCR. The composition of microbiota between UC and non-IBD differed significantly (PCoA, p = 0.001). UC children had a decrease in Bacteroidetes and an increase in several family-level taxa including Peptostreptococcaceae and Enterobacteriaceae, which correlated negatively with the expression of antimicrobial peptides REG3G and DEFB1, respectively. Enterobacteriaceae correlated positively with the expression siderophore binding protein LCN2 and Betaproteobacteria negatively with DEFB4A expression. The results indicate that reciprocal interaction of epithelial microbiota and defense mechanisms play a role in UC.


Subject(s)
Colitis, Ulcerative/microbiology , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/physiology , Pore Forming Cytotoxic Proteins/genetics , Adolescent , Bacteroidetes/genetics , Case-Control Studies , Child , Child, Preschool , DNA, Ribosomal , Enterobacteriaceae/genetics , Finland , Gastrointestinal Microbiome/genetics , Gene Expression , Humans , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/pathology , Lipocalin-2/genetics , Pancreatitis-Associated Proteins/genetics , beta-Defensins/genetics
5.
Nutrients ; 12(9)2020 Aug 25.
Article in English | MEDLINE | ID: mdl-32854216

ABSTRACT

A gluten-free diet may result in high fat and low fiber intake and thus lead to unbalanced microbiota. This study characterized fecal microbiota profiles by 16S MiSeq sequencing among oat-using healthy adult subjects (n = 14) or adult subjects with celiac disease (CeD) (n = 19) or non-celiac gluten sensitivity (NCGS) (n = 10). Selected microbial metabolites, self-reported 4d food diaries and perceived gut symptoms were compared. Subjects with NCGS experienced the highest amount of gut symptoms and received more energy from fat and less from carbohydrates than healthy and CeD subjects. Oat consumption resulted in reaching the lower limit of the recommended fiber intake. Frequent consumption of gluten-free pure oats did not result in microbiota dysbiosis in subjects with CeD or NCGS. Thus, the high number of gut symptoms in NCGS subjects was not linked to the microbiota. The proportion of fecal acetate was higher in healthy when compared to NCGS subjects, which may be linked to a higher abundance of Bifidobacterium in the control group compared to NCGS and CeD subjects. Propionate, butyrate and ammonia production and ß-glucuronidase activity were comparable among the study groups. The results suggest that pure oats have great potential as the basis of a gluten-free diet and warrant further studies in minor microbiota disorders.


Subject(s)
Avena , Celiac Disease/microbiology , Edible Grain , Feces/microbiology , Gastrointestinal Microbiome , Glutens , Adult , Aged , Bacteria/metabolism , Celiac Disease/metabolism , Diet Records , Diet, Gluten-Free , Dietary Carbohydrates/administration & dosage , Dietary Fats/administration & dosage , Dietary Fiber/administration & dosage , Fatty Acids, Volatile/metabolism , Female , Glutens/metabolism , Humans , Male , Middle Aged , Young Adult
6.
Ageing Res Rev ; 62: 101079, 2020 09.
Article in English | MEDLINE | ID: mdl-32461136

ABSTRACT

The global increases in life expectancy and population have resulted in a growing ageing population and with it a growing number of people living with age-related neurodegenerative conditions and dementia, shifting focus towards methods of prevention, with lifestyle approaches such as nutrition representing a promising avenue for further development. This overview summarises the main themes discussed during the 3rd Symposium on "Nutrition for the Ageing Brain: Moving Towards Clinical Applications" held in Madrid in August 2018, enlarged with the current state of knowledge on how nutrition influences healthy ageing and gives recommendations regarding how the critical field of nutrition and neurodegeneration research should move forward into the future. Specific nutrients are discussed as well as the impact of multi-nutrient and whole diet approaches, showing particular promise to combatting the growing burden of age-related cognitive decline. The emergence of new avenues for exploring the role of diet in healthy ageing, such as the impact of the gut microbiome and development of new techniques (imaging measures of brain metabolism, metabolomics, biomarkers) are enabling researchers to approach finding answers to these questions. But the translation of these findings into clinical and public health contexts remains an obstacle due to significant shortcomings in nutrition research or pressure on the scientific community to communicate recommendations to the general public in a convincing and accessible way. Some promising programs exist but further investigation to improve our understanding of the mechanisms by which nutrition can improve brain health across the human lifespan is still required.


Subject(s)
Healthy Aging , Nutritional Status , Aging , Brain , Diet , Humans
7.
Clin Nutr ; 39(9): 2647-2662, 2020 09.
Article in English | MEDLINE | ID: mdl-32035751

ABSTRACT

BACKGROUND & AIMS: Aberrations in body composition are expected in children suffering from chronic inflammatory conditions. The objective is to examine whether children with inflammatory bowel disease (IBD: Crohn's disease and ulcerative colitis), coeliac disease, asthma and juvenile idiopathic arthritis (JIA) have an altered body composition as compared to healthy children. METHODS: A systematic review, registered in Prospero (registration number: CRD42018107645), was conducted according to PRISMA guidelines. We conducted a search of three databases, Pubmed, Cochrane and Scopus. An assessment of the quality of the study was performed. RESULTS: Data from 50 studies, 32 with IBD, 8 with coeliac disease, 2 with asthma and 8 with JIA, involving 2399 children were selected for review after applying the eligibility criteria. In all but 4 studies, children with Crohn's disease exhibited decreased amounts of fat mass and fat free mass. Reductions in fat mass were also evident in studies in children with coeliac disease. It is uncertain whether body composition is altered in children with asthma or JIA. CONCLUSIONS: Children with Crohn's disease manifest with lowered adiposity and lean mass and therefore are likely to be at risk for suffering malnutrition-related clinical complications. Apart from Crohn's disease, data examining body composition in children with chronic inflammatory conditions are scarce and there is a paucity of reports examining the relationship between inflammation and body composition. Interpretation of the current study results is hampered by the low quality of the studies and due to the fact that the analyses have been habitually secondary outcomes.


Subject(s)
Arthritis, Juvenile/physiopathology , Asthma/physiopathology , Body Composition/physiology , Celiac Disease/physiopathology , Colitis, Ulcerative/physiopathology , Crohn Disease/physiopathology , Adiposity/physiology , Adolescent , Child , Child, Preschool , Female , Humans , Inflammation/physiopathology , Male , Nutritional Status
8.
Nutrients ; 10(12)2018 Nov 29.
Article in English | MEDLINE | ID: mdl-30501103

ABSTRACT

The potential association between gut microbiota perturbations and childhood functional gastrointestinal disturbances opens interesting therapeutic and preventive possibilities with probiotics. The aim of this review was to evaluate current evidence on the efficacy of probiotics for the management of pediatric functional abdominal pain disorders, functional constipation and infantile colic. Thus far, no single strain, combination of strains or synbiotics can be recommended for the management of irritable bowel syndrome, functional abdominal pain or functional constipation in children. However, Lactobacillus reuteri DSM 17938 may be considered for the management of breastfed colic infants, while data on other probiotic strains, probiotic mixtures or synbiotics are limited in infantile colic.


Subject(s)
Gastrointestinal Diseases , Probiotics/therapeutic use , Abdominal Pain , Adolescent , Child , Child, Preschool , Colic , Humans , Infant , Infant, Newborn
9.
Neonatology ; 114(3): 226-229, 2018.
Article in English | MEDLINE | ID: mdl-29940563

ABSTRACT

BACKGROUND: The aetiology of infantile colic remains unknown. However, altered gut microbiota composition has been reported in children with the disorder. OBJECTIVE: The objective of this study was to determine the associations between perinatal factors potentially affecting gut colonization and infantile colic. METHODS: Altogether 48 infants with colic and 29 controls were selected from 2 ongoing clinical studies. Infants with and without colic were comparable with regard to their background characteristics. RESULTS: A significant difference was detected in intrapartum antibiotic use and breastfeeding rates between infants with and without colic. The association between exposure to intrapartum antibiotics and infantile colic remained statistically significant after adjusting for potential confounding factors. CONCLUSIONS: Since intrapartum antibiotic exposure may have an effect on early gut colonization, our finding is consistent with the association between aberrant early gut microbiota composition and development of colic. Antibiotic-exposed neonates may represent a novel target group for preventive intervention studies.


Subject(s)
Anti-Bacterial Agents/adverse effects , Breast Feeding , Colic/chemically induced , Gastrointestinal Microbiome/drug effects , Maternal Exposure/adverse effects , Anti-Bacterial Agents/administration & dosage , Case-Control Studies , Female , Humans , Infant , Infant, Newborn , Logistic Models , Male , Parturition
11.
J Pediatr Gastroenterol Nutr ; 64(5): 691-695, 2017 05.
Article in English | MEDLINE | ID: mdl-27478897

ABSTRACT

OBJECTIVES: Dysbiosis, an imbalance in the taxonomic composition of the gut bacteria occurring during the critical stages of development, induces lasting shifts in the immunological and metabolic phenotype if accompanied by an inflammatory response. Because altered gut microbiota and successful treatment with probiotics have both been demonstrated in cases of colic, we hypothesized here that infants with colic might have low-grade inflammation. METHODS: In 28 infants with colic and in 12 healthy controls at the age of 1 month, we measured the following serum immunological biomarkers: cytokines interleukin 1ß (IL-1ß); IL-6; IL-10; tumor necrosis factor α; interferon γ (IFN-γ); chemokines IL-8; monocyte chemotactic protein-1 (MCP-1); macrophage inflammatory protein 1ß (MIP-1ß) and chemokine (C-X-C motif) ligand 16; and intestinal fatty acid-binding protein, a biomarker of enterocyte damage and zonulin, a biomarker of intestinal permeability. In addition, intestinal microbiota composition was correlated with immunological biomarkers. RESULTS: Infants with colic had increased concentrations of IL-8, MCP-1, and MIP-1ß in serum as compared with healthy children. All the other immunological biomarkers were comparable between the groups. Fecal levels of Clostridium leptum correlated negatively with the proinflammatory markers MCP-1 (r = -0.44, P = 0.02), MIP-1ß (r = -0.43, P = 0.02), and tumor necrosis factor α (r = -0.38, P = 0.04). In addition, C coccoides group levels correlated negatively with MCP-1 (r = -0.43, P = 0.02) and Bifidobacterium breve levels positively with chemokine (C-X-C motif) ligand 16 (r = 0.38, P = 0.04). CONCLUSIONS: In addition to gut microbiota alterations, colic in infants is associated with low-grade systemic inflammation. Specific bacterial species beyond conventional probiotics may have anti-inflammatory properties that may help to modulate microbiota and alleviate colic-related inflammation.


Subject(s)
Colic/immunology , Cytokines/blood , Inflammation/complications , Biomarkers/blood , Case-Control Studies , Colic/microbiology , Feces/microbiology , Female , Gastrointestinal Microbiome/immunology , Humans , Infant , Inflammation/blood , Inflammation/diagnosis , Inflammation/microbiology , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/microbiology , Male , Permeability , Prospective Studies
12.
Front Microbiol ; 7: 1706, 2016.
Article in English | MEDLINE | ID: mdl-27833600

ABSTRACT

Sutterella species have been frequently associated with human diseases, such as autism, Down syndrome, and inflammatory bowel disease (IBD), but the impact of these bacteria on health still remains unclear. Especially the interactions of Sutterella spp. with the host are largely unknown, despite of the species being highly prevalent. In this study, we addressed the interaction of three known species of Sutterella with the intestinal epithelium and examined their adhesion properties, the effect on intestinal barrier function and the pro-inflammatory capacity in vitro. We also studied the relative abundance and prevalence of the genus Sutterella and Sutterella wadsworthensis in intestinal biopsies of healthy individuals and patients with celiac disease (CeD) or IBD. Our results show that Sutterella spp. are abundant in the duodenum of healthy adults with a decreasing gradient toward the colon. No difference was detected in the prevalence of Sutterella between the pediatric IBD or CeD patients and the healthy controls. Sutterella parvirubra adhered better than the two other Sutterella spp. to differentiated Caco-2 cells and was capable of decreasing the adherence of S. wadsworthensis, which preferably bound to mucus and human extracellular matrix proteins. Furthermore, only S. wadsworthensis induced an interleukin-8 production in enterocytes, which could be due to different lipopolysaccharide structures between the species. However, its pro-inflammatory activity was modest as compared to non-pathogenic Escherichia coli. Sutterella spp. had no effect on the enterocyte monolayer integrity in vitro. Our findings indicate that the members of genus Sutterella are widely prevalent commensals with mild pro-inflammatory capacity in the human gastrointestinal tract and do not contribute significantly to the disrupted epithelial homeostasis associated with microbiota dysbiosis and increase of Proteobacteria. The ability of Sutterella spp. to adhere to intestinal epithelial cells indicate that they may have an immunomodulatory role.

13.
Pediatr Res ; 78(4): 470-5, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26151493

ABSTRACT

BACKGROUND: Probiotic Lactobacillus reuteri and reduced allergen load may lessen the daily crying of colic infants, but the role of Lactobacillus rhamnosus GG (LGG) has remained obscure. METHODS: Infants with colic (n = 30) were enrolled during the first 6 wk of life. All families received behavioral support and allergen avoidance diet: breastfeeding mothers followed cow's milk elimination diet and formula-fed infants received extensively hydrolyzed casein formula. The randomized, double-blind intervention employed of LGG 4.5 × 10(9) cfu/d or placebo for a 4-wk study period. Daily crying was recorded by diaries and parental interviews. Fecal calprotectin and gut microbiota composition by quantitative PCR were evaluated before and after the intervention. RESULTS: Daily crying time was comparable between the probiotic (173 min) and the placebo group (174 min; P = 0.99) at the end of the intervention according to the parental diary. However, parents reported a decrease of 68% (95% confidence interval (CI): 58-78) in daily crying in the probiotic and 49% (95% CI: 32-66) in the placebo group (P = 0.05). CONCLUSION: LGG in infants treated in tandem with behavioral support and a cow's milk elimination diet did not provide additional treatment effect for diary-verified colic crying although parental report of crying suggested the probiotic intervention effective.


Subject(s)
Colic/therapy , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Lacticaseibacillus rhamnosus/growth & development , Probiotics/therapeutic use , Behavior Therapy , Bottle Feeding , Breast Feeding , Caseins/administration & dosage , Colic/diagnosis , Colic/microbiology , Colic/psychology , Combined Modality Therapy , Crying , DNA, Bacterial/genetics , Double-Blind Method , Female , Finland , Humans , Infant , Infant Behavior , Infant Formula , Infant, Newborn , Interviews as Topic , Lacticaseibacillus rhamnosus/genetics , Male , Polymerase Chain Reaction , Time Factors , Treatment Outcome
14.
Pediatr Res ; 77(6): 823-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25760553

ABSTRACT

BACKGROUND: Recent experimental evidence suggests that gut microbiota may alter function within the nervous system providing new insight on the mechanism of neuropsychiatric disorders. METHODS: Seventy-five infants who were randomized to receive Lactobacillus rhamnosus GG (ATCC 53103) or placebo during the first 6 mo of life were followed-up for 13 y. Gut microbiota was assessed at the age of 3 wk, 3, 6, 12, 18, 24 mo, and 13 y using fluorescein in situ hybridization (FISH) and qPCR, and indirectly by determining the blood group secretor type at the age of 13 y. The diagnoses of attention deficit hyperactivity disorder (ADHD) and Asperger syndrome (AS) by a child neurologist or psychiatrist were based on ICD-10 diagnostic criteria. RESULTS: At the age of 13 y, ADHD or AS was diagnosed in 6/35 (17.1%) children in the placebo and none in the probiotic group (P = 0.008). The mean (SD) numbers of Bifidobacterium species bacteria in feces during the first 6 mo of life was lower in affected children 8.26 (1.24) log cells/g than in healthy children 9.12 (0.64) log cells/g; P = 0.03. CONCLUSION: Probiotic supplementation early in life may reduce the risk of neuropsychiatric disorder development later in childhood possible by mechanisms not limited to gut microbiota composition.


Subject(s)
Asperger Syndrome/prevention & control , Attention Deficit Disorder with Hyperactivity/prevention & control , Gastrointestinal Microbiome/genetics , Probiotics/pharmacology , Adolescent , Age Factors , Bifidobacterium/isolation & purification , Dietary Supplements , Feces/microbiology , Humans , In Situ Hybridization, Fluorescence , Infant , Polymerase Chain Reaction , Probiotics/administration & dosage
15.
J Crohns Colitis ; 8(10): 1222-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24630488

ABSTRACT

BACKGROUND AND AIMS: CXCL16 is a scavenger receptor which has been connected to phagocytosis of bacterial antigens in experimental colitis. It has also been shown to have a pivotal role in the development of experimental colitis in mice. The increased expression of CXCL16 has been demonstrated in inflamed lesions of patients with Crohn disease. Our aim was to study the expression of CXCL16 in the colon of patients with ulcerative colitis. METHODS: Relative quantitative reverse transcription-polymerase chain reaction was applied to explore the gene expressions of CXCL16, its receptor CXCR6, and interleukin 8, an inflammatory marker, in the colonic biopsies of children with active ulcerative colitis (n=19), children with ulcerative colitis in remission (n=9) and children with no inflammatory condition in colon (n=14). RESULTS: An increased expression of CXCL16 in the colonic biopsies of children with ulcerative colitis was found both in active disease (p=0.006) and in remission (p=0.033), when compared to children without inflammatory condition. The gene expressions of interleukin 8 and CXCL16 correlated with each other (rs=0.67, p=0.01). The expression of CXCR6 mRNA was comparable between the study groups (p=0.50). CONCLUSIONS: The gene expression of CXCL16 was increased in patients with ulcerative colitis both in active disease and in remission suggesting an important role of the molecule in the pathogenesis of the condition.


Subject(s)
Chemokines, CXC/genetics , Colitis, Ulcerative/genetics , Interleukin-8/genetics , RNA, Messenger/genetics , Receptors, Chemokine/genetics , Receptors, Scavenger/genetics , Receptors, Virus/genetics , Adolescent , Chemokine CXCL16 , Child , Child, Preschool , Gene Expression , Humans , Receptors, CXCR6
16.
J Allergy Clin Immunol ; 133(2): 405-13, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24131826

ABSTRACT

BACKGROUND: Simple and safe strategies for the prevention of viral respiratory tract infections (RTIs) are needed. OBJECTIVE: We hypothesized that early prebiotic or probiotic supplementation would reduce the risk of virus-associated RTIs during the first year of life in a cohort of preterm infants. METHODS: In this randomized, double-blind, placebo-controlled trial (ClinicalTrials.gov no. NCT00167700), 94 preterm infants (gestational age, ≥32 + 0 and ≤36 + 6 weeks; birth weight, >1500 g) treated at Turku University Hospital, Turku, Finland, were allocated to receive oral prebiotics (galacto-oligosaccharide and polydextrose mixture, 1:1), a probiotic (Lactobacillus rhamnosus GG, ATCC 53103), or placebo (microcrystalline cellulose) between days 3 and 60 of life. The primary outcome was the incidence of clinically defined virus-associated RTI episodes confirmed from nasal swabs by using nucleic acid testing. Secondary outcomes were the severity and duration of RTIs. RESULTS: A significantly lower incidence of RTIs was detected in infants receiving prebiotics (rate ratio [RR], 0.24; 95% CI, 0.12-0.49; P < .001) or probiotics (RR, 0.50; 95% CI, 0.28-0.90; P = .022) compared with those receiving placebo. Also, the incidence of rhinovirus-induced episodes, which comprised 80% of all RTI episodes, was found to be significantly lower in the prebiotic (RR, 0.31; 95% CI, 0.14-0.66; P = .003) and probiotic (RR, 0.49; 95% CI, 0.24-1.00; P = .051) groups compared with the placebo group. No differences emerged among the study groups in rhinovirus RNA load during infections, duration of rhinovirus RNA shedding, duration or severity of rhinovirus infections, or occurrence of rhinovirus RNA in asymptomatic infants. CONCLUSIONS: Gut microbiota modification with specific prebiotics and probiotics might offer a novel and cost-effective means to reduce the risk of rhinovirus infections.


Subject(s)
Picornaviridae Infections/prevention & control , Prebiotics , Probiotics/therapeutic use , Respiratory Tract Diseases/prevention & control , Rhinovirus/physiology , DNA, Viral/analysis , Double-Blind Method , Female , Humans , Infant , Infant, Newborn , Infant, Premature , Male , Picornaviridae Infections/virology , Respiratory Tract Diseases/virology , Viral Load
18.
J Pediatr ; 163(5): 1272-7.e1-2, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23915796

ABSTRACT

OBJECTIVE: To evaluate the impact of early prebiotic and probiotic intervention on preterm infants' well-being, crying, growth, and microbiological programming. STUDY DESIGN: Ninety-four preterm infants (gestational age 32-36 weeks and birth weight >1500 g) randomized to receive prebiotics (mixture of galacto-oligosaccharide and polydextrose 1:1), probiotics (Lactobacillus rhamnosus GG), or placebo during the first 2 months of life were followed up for 1 year. Infants were categorized based on the extent of crying and irritability during the first 2 months of life, and their gut microbiota was investigated by fluorescence in situ hybridization (n = 66) and quantitative polymerase chain reaction (n = 63). RESULTS: A total of 27 of 94 infants (29%) infants were classified as excessive criers, significantly less frequently in the prebiotic and the probiotic groups than in the placebo group (19% vs 19% vs 47%, respectively; P = .02). The placebo group had a higher percentage of Clostridium histolyticum group bacteria in their stools than did the probiotic group (13.9% vs 8.9%, respectively; P = .05). There were no adverse events related to either supplementation. CONCLUSIONS: Early prebiotic and probiotic supplementation may alleviate symptoms associated with crying and fussing in preterm infants. This original finding may offer new therapeutic and preventive measures for this common disturbance in early life.


Subject(s)
Dietary Supplements , Infant, Premature/growth & development , Intestines/microbiology , Prebiotics , Probiotics/administration & dosage , Bifidobacterium/genetics , Crying , Double-Blind Method , Female , Follow-Up Studies , Galactose/chemistry , Gestational Age , Glucans/administration & dosage , Humans , In Situ Hybridization, Fluorescence , Infant , Infant, Newborn , Lacticaseibacillus rhamnosus/metabolism , Male , Oligosaccharides/administration & dosage
19.
BMC Gastroenterol ; 13: 113, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23844808

ABSTRACT

BACKGROUND: Celiac disease (CD) is an autoimmune disorder of the small intestine which is triggered by dietary gluten in genetically predisposed (HLA-DQ2/DQ8 positive) individuals. Only a fraction of HLA-DQ2/DQ8 positive individuals develop CD indicating that other factors have a role in the disorder. Several studies have addressed intestinal microbiota aberrancies in pediatric CD, but the results are inconsistent. Previously, we demonstrated that pediatric CD patients have lower duodenal expression of TLR2 and higher expression of TLR9 as compared to healthy controls (HC) indicating that microbiota may have a role in CD. METHODS: We used bacterial phylogenetic microarray to comprehensively profile the microbiota in duodenal biopsies of CD (n = 10) and HC (n = 9) children. The expression of selected mucosa-associated genes was assessed by qRT-PCR in CD and HC children and in treated CD adults (T-CD, n = 6) on gluten free diet. RESULTS: The overall composition, diversity and the estimated microbe associated molecular pattern (MAMP) content of microbiota were comparable between CD and HC, but a sub-population profile comprising eight genus-like bacterial groups was found to differ significantly between HC and CD. In HC, increased TLR2 expression was positively correlated with the expression of tight junction protein ZO-1. In CD and T-CD, the expression of IL-10, IFN-g and CXCR6 were higher as compared to HC. CONCLUSIONS: The results suggest that microbiota and altered expression of mucosal receptors have a role in CD. In CD subjects, the increased expression of IL-10 and IFN-g may have partly resulted from the increased TLR9 expression and signaling.


Subject(s)
Celiac Disease/metabolism , Celiac Disease/microbiology , Duodenum/metabolism , Duodenum/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Adolescent , Adult , Case-Control Studies , Celiac Disease/genetics , Chemokine CXCL16 , Chemokines, CXC/genetics , Child , Child, Preschool , Connexin 43/genetics , Female , Gene Expression , Homeostasis , Humans , Interferon-gamma/genetics , Interleukin-10/genetics , Male , Metagenome , Middle Aged , Mucin-2/genetics , Pancreatitis-Associated Proteins , Proteins/genetics , Receptors, CXCR6 , Receptors, Chemokine/genetics , Receptors, Scavenger/genetics , Receptors, Virus/genetics , Signal Transduction , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 9/metabolism , Tumor Necrosis Factor-alpha/genetics , Zonula Occludens-1 Protein/genetics
20.
BMC Microbiol ; 13: 12, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23339708

ABSTRACT

BACKGROUND: Deviations in composition and diversity of intestinal microbiota in infancy have been associated with both the development and recurrence of atopic eczema. Thus, we decided to use a deep and global microarray-based method to characterize the diversity and temporal changes of the intestinal microbiota in infancy and to define specific bacterial signatures associated with eczema. Faecal microbiota at 6 and 18 months of age were analysed from 34 infants (15 with eczema and 19 healthy controls) selected from a prospective follow-up study based on the availability of faecal samples. The infants were originally randomized to receive either Lactobacillus rhamnosus GG or placebo. RESULTS: Children with eczema harboured a more diverse total microbiota than control subjects as assessed by the Simpson's reciprocal diversity index of the microarray profiles. Composition of the microbiota did not differ between study groups at age of 6 months, but was significantly different at age of 18 months as assessed by MCPP (p=0.01). At this age healthy children harboured 3 -fold greater amount of members of the Bacteroidetes (p=0.01). Microbiota of children suffering from eczema had increased abundance of the Clostridium clusters IV and XIVa, which are typically abundant in adults. Probiotic Lactobacillus rhamnosus GG supplementation in early infancy was observed to have minor long-term effects on the microbiota composition. CONCLUSION: A diverse and adult-type microbiota in early childhood is associated with eczema and it may contribute to the perpetuation of eczema.


Subject(s)
Bacteria/classification , Bacteria/genetics , Biodiversity , Dermatitis, Atopic/microbiology , Gastrointestinal Tract/microbiology , Metagenome , Microarray Analysis , Female , Follow-Up Studies , Humans , Infant , Male , Placebos/administration & dosage , Probiotics/administration & dosage , Randomized Controlled Trials as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...