Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Allergy Clin Immunol ; 149(2): 650-658.e5, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34224785

RESUMEN

BACKGROUND: Tolerance development is an important clinical outcome for infants with cow's milk allergy. OBJECTIVE: This multicenter, prospective, randomized, double-blind, controlled clinical study (NTR3725) evaluated tolerance development to cow's milk (CM) and safety of an amino acid-based formula (AAF) including synbiotics (AAF-S) comprising prebiotic oligosaccharides (oligofructose, inulin) and probiotic Bifidobacterium breve M-16V in infants with confirmed IgE-mediated CM allergy. METHODS: Subjects aged ≤13 months with IgE-mediated CM allergy were randomized to receive AAF-S (n = 80) or AAF (n = 89) for 12 months. Stratification was based on CM skin prick test wheal size and study site. After 12 and 24 months, CM tolerance was evaluated by double-blind, placebo-controlled food challenge. A logistic regression model used the all-subjects randomized data set. RESULTS: At baseline, mean ± SD age was 9.36 ± 2.53 months. At 12 and 24 months, respectively, 49% and 62% of subjects were CM tolerant (AAF-S 45% and 64%; AAF 52% and 59%), and not differ significantly between groups. During the 12-month intervention, the number of subjects reporting at least 1 adverse event did not significantly differ between groups; however, fewer subjects required hospitalization due to serious adverse events categorized as infections in the AAF-S versus AAF group (9% vs 20%; P = .036). CONCLUSIONS: After 12 and 24 months, CM tolerance was not different between groups and was in line with natural outgrowth. Results suggest that during the intervention, fewer subjects receiving AAF-S required hospitalization due to infections.


Asunto(s)
Aminoácidos/administración & dosificación , Tolerancia Inmunológica , Fórmulas Infantiles , Hipersensibilidad a la Leche/inmunología , Método Doble Ciego , Femenino , Humanos , Lactante , Fórmulas Infantiles/efectos adversos , Recién Nacido , Masculino , Estudios Prospectivos , Simbióticos/administración & dosificación
2.
Gut ; 71(9): 1803-1811, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-34803023

RESUMEN

OBJECTIVE: Revised guidelines for caesarean section (CS) advise maternal antibiotic administration prior to skin incision instead of after umbilical cord clamping, unintentionally exposing the infant to antibiotics antenatally. We aimed to investigate if timing of intrapartum antibiotics contributes to the impairment of microbiota colonisation in CS born infants. DESIGN: In this randomised controlled trial, women delivering via CS received antibiotics prior to skin incision (n=20) or after umbilical cord clamping (n=20). A third control group of vaginally delivering women (n=23) was included. Faecal microbiota was determined from all infants at 1, 7 and 28 days after birth and at 3 years by 16S rRNA gene sequencing and whole-metagenome shotgun sequencing. RESULTS: Compared with vaginally born infants, profound differences were found in microbial diversity and composition in both CS groups in the first month of life. A decreased abundance in species belonging to the genera Bacteroides and Bifidobacterium was found with a concurrent increase in members belonging to the phylum Proteobacteria. These differences could not be observed at 3 years of age. No statistically significant differences were observed in taxonomic and functional composition of the microbiome between both CS groups at any of the time points. CONCLUSION: We confirmed that microbiome colonisation is strongly affected by CS delivery. Our findings suggest that maternal antibiotic administration prior to CS does not result in a second hit on the compromised microbiome. Future, larger studies should confirm that antenatal antibiotic exposure in CS born infants does not aggravate colonisation impairment and impact long-term health.


Asunto(s)
Antibacterianos , Cesárea , Antibacterianos/uso terapéutico , Bacteroides , Bifidobacterium , Cesárea/efectos adversos , Heces/microbiología , Femenino , Humanos , Lactante , Embarazo , ARN Ribosómico 16S/genética
3.
Am J Physiol Gastrointest Liver Physiol ; 322(6): G571-G582, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35348015

RESUMEN

This study examined fecal metabolome dynamics to gain greater functional insights into the interactions between nutrition and the activity of the developing gut microbiota in healthy term-born infants. The fecal samples used here originate from a randomized, controlled, double-blind clinical study that assessed the efficacy of infant formula with prebiotics and postbiotics (experimental arm) compared with a standard infant formula (control arm). A group of exclusively breast-fed term infants was used as a reference arm. First, conventional targeted physiological and microbial measurements were performed, which showed differences in fecal Bifidobacterium levels and corresponding activity (e.g., lactate levels). Next, the overall fecal microbiota composition was determined by 16S rRNA gene amplicon sequencing. The microbiota composition profiles showed several bacterial groups in the experimental arm to be significantly different from the control arm and mostly closer to the levels observed in the reference arm. Finally, we applied an untargeted UPLC-MS/MS approach to examine changes in the fecal metabolome. Fecal metabolome profiles showed the most distinct separation, up to 404 significantly different metabolites, between the study arms. Our data reveal that infant formula with specific prebiotics and postbiotics may trigger responses in the intestinal microbiota composition that brings the ensuing fecal metabolite profile of formula-fed infants closer toward those observed in breast-fed infants. Furthermore, our results demonstrate a clear need for establishing an infant gut metabolome reference database to translate these metabolite profile dynamics into functional and physiologically relevant responses.NEW & NOTEWORTHY Untargeted metabolomics techniques can provide a "snapshot" of an ecosystem in response to environmental stimuli, such as nutritional interventions. Our analyses of fecal samples from infants demonstrate the potential of phenotyping by metabolomics while deciphering the complex interactions of early-life nutrition and gut microbiome development.


Asunto(s)
Fórmulas Infantiles , Microbiota , Cromatografía Liquida , Heces/química , Femenino , Humanos , Lactante , Metaboloma , Prebióticos , ARN Ribosómico 16S , Espectrometría de Masas en Tándem
4.
Mol Genet Metab ; 137(3): 308-322, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36274442

RESUMEN

Propionic acidemia (PA) is an inherited metabolic disorder of propionate metabolism, where the gut microbiota may play a role in pathophysiology and therefore, represent a relevant therapeutic target. Little is known about the gut microbiota composition and activity in patients with PA. Although clinical practice varies between metabolic treatment centers, management of PA requires combined dietary and pharmaceutical treatments, both known to affect the gut microbiota. This study aimed to characterize the gut microbiota and its metabolites in fecal samples of patients with PA compared with healthy controls from the same household. Eight patients (aged 3-14y) and 8 controls (4-31y) were recruited from Center 1 (UK) and 7 patients (11-33y) and 6 controls (15-54y) from Center 2 (Austria). Stool samples were collected 4 times over 3 months, alongside data on dietary intakes and medication usage. Several microbial taxa differed between patients with PA and controls, particularly for Center 1, e.g., Proteobacteria levels were increased, whereas butyrate-producing genera, such as Roseburia and Faecalibacterium, were decreased. Most measured microbial metabolites were lower in patients with PA, and butyrate was particularly depleted in patients from Center 1. Furthermore, microbiota profile of these patients showed the lowest compositional and functional diversity, and lowest stability over 3 months. As the first study to map the gut microbiota of patients with PA, this work represents an important step forward for developing new therapeutic strategies to further improve PA clinical status. New dietary strategies should consider microbial propionate production as well as butyrate production and microbiota stability.


Asunto(s)
Microbioma Gastrointestinal , Acidemia Propiónica , Humanos , Propionatos , Heces/microbiología , Butiratos
5.
Environ Microbiol ; 23(12): 7710-7722, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34309161

RESUMEN

Exposure to a diverse microbial environment during pregnancy and early postnatal period is important in determining predisposition towards allergy. However, the effect of environmental microbiota exposure during preconception, pregnancy and postnatal life on development of allergy in the child has not been investigated so far. In the S-PRESTO (Singapore PREconception Study of long Term maternal and child Outcomes) cohort, we collected house dust during all three critical window periods and analysed microbial composition using 16S rRNA gene sequencing. At 6 and 18 months, the child was assessed for eczema by clinicians. In the eczema group, household environmental microbiota was characterized by presence of human-associated bacteria Actinomyces, Anaerococcus, Finegoldia, Micrococcus, Prevotella and Propionibacterium at all time points, suggesting their possible contributions to regulating host immunity and increasing the susceptibility to eczema. In the home environment of the control group, putative protective effect of an environmental microbe Planomicrobium (Planococcaceae family) was observed to be significantly higher than that in the eczema group. Network correlation analysis demonstrated inverse relationships between beneficial Planomicrobium and human-associated bacteria (Actinomyces, Anaerococcus, Finegoldia, Micrococcus, Prevotella and Propionibacterium). Exposure to natural environmental microbiota may be beneficial to modulate shed human-associated microbiota in an indoor environment.


Asunto(s)
Eccema , Microbiota , Bacterias/genética , Niño , Estudios de Cohortes , Femenino , Humanos , Microbiota/genética , Embarazo , ARN Ribosómico 16S/genética
6.
BMC Microbiol ; 21(1): 191, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34172012

RESUMEN

BACKGROUND: The compromised gut microbiome that results from C-section birth has been hypothesized as a risk factor for the development of non-communicable diseases (NCD). In a double-blind randomized controlled study, 153 infants born by elective C-section received an infant formula supplemented with either synbiotic, prebiotics, or unsupplemented from birth until 4 months old. Vaginally born infants were included as a reference group. Stool samples were collected from day 3 till week 22. Multi-omics were deployed to investigate the impact of mode of delivery and nutrition on the development of the infant gut microbiome, and uncover putative biological mechanisms underlying the role of a compromised microbiome as a risk factor for NCD. RESULTS: As early as day 3, infants born vaginally presented a hypoxic and acidic gut environment characterized by an enrichment of strict anaerobes (Bifidobacteriaceae). Infants born by C-section presented the hallmark of a compromised microbiome driven by an enrichment of Enterobacteriaceae. This was associated with meta-omics signatures characteristic of a microbiome adapted to a more oxygen-rich gut environment, enriched with genes associated with reactive oxygen species metabolism and lipopolysaccharide biosynthesis, and depleted in genes involved in the metabolism of milk carbohydrates. The synbiotic formula modulated expression of microbial genes involved in (oligo)saccharide metabolism, which emulates the eco-physiological gut environment observed in vaginally born infants. The resulting hypoxic and acidic milieu prevented the establishment of a compromised microbiome. CONCLUSIONS: This study deciphers the putative functional hallmarks of a compromised microbiome acquired during C-section birth, and the impact of nutrition that may counteract disturbed microbiome development. TRIAL REGISTRATION: The study was registered in the Dutch Trial Register (Number: 2838 ) on 4th April 2011.


Asunto(s)
Bacterias/genética , Cesárea/efectos adversos , Heces/microbiología , Microbioma Gastrointestinal/genética , Metagenoma/genética , Biodiversidad , Método Doble Ciego , Humanos , Lactante , Recién Nacido
7.
Nutr Neurosci ; 23(11): 896-910, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30871432

RESUMEN

Objective: The intestinal microbiota is acknowledged to be essential in brain development and behaviour. Their composition can be modulated by prebiotics such as short-chain galacto-oligosaccharides (scGOS) and long-chain fructo-oligosaccharide (lcFOS). Several studies reported potential health benefit of prebiotics on behaviour. As the prebiotic mixture of scGOS and lcFOS is included in infant formula, we investigated the effects of dietary supplementation with this specific mixture from the day of birth onwards on behaviour and intestinal microbiota development in mice. Method: Healthy male BALB/cByJ mice received, from day of birth, a dietary supplement with or without 3% scGOS:lcFOS (9:1). Behavioural tests were performed pre-weaning, in adolescence, early adulthood and adulthood. We assessed faecal microbiota compositions over time, caecal short-chain fatty acids as well as brain mRNA expression of Htr1a, Htr1b and Tph2 and monoamine levels. Results: Compared to control fed mice, scGOS:lcFOS fed mice showed reduced anxiety-like and repetitive behaviour over time and improved social behaviour in adulthood. The serotonergic system in the prefrontal cortex (PFC) and somatosensory cortex (SSC) was affected by the scGOS:lcFOS. In the PFC, mRNA expression of brain-derived neurotrophic factor (Bdnf) was enhanced in scGOS:lcFOS fed mice. Although the bacterial diversity of the intestinal microbiota was unaffected by the scGOS:lcFOS diet, microbiota composition differed between the scGOS:lcFOS and the control fed mice over time. Moreover, an increased saccharolytic and decreased proteolytic fermentation activity were observed in caecum content. Discussion: Supplementing the diet with scGOS:lcFOS from the day of birth is associated with reduced anxiety-like and improved social behaviour during the developmental period and later in life, and modulates the composition and activity of the intestinal microbiota in healthy male BALB/c mice. These data provide further evidence of the potential impact of scGOS:lcFOS on behaviour at several developmental stages throughout life and strengthen the insights in the interplay between the developing intestine and brain.


Asunto(s)
Ansiedad/microbiología , Microbioma Gastrointestinal , Oligosacáridos/administración & dosificación , Prebióticos/administración & dosificación , Conducta Social , Animales , Ansiedad/prevención & control , Conducta Animal , Monoaminas Biogénicas/metabolismo , Encéfalo/metabolismo , Masculino , Ratones Endogámicos BALB C , Vocalización Animal
8.
Nucleic Acids Res ; 46(4): 1860-1877, 2018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29294107

RESUMEN

Bifidobacterium breve represents one of the most abundant bifidobacterial species in the gastro-intestinal tract of breast-fed infants, where their presence is believed to exert beneficial effects. In the present study whole genome sequencing, employing the PacBio Single Molecule, Real-Time (SMRT) sequencing platform, combined with comparative genome analysis allowed the most extensive genetic investigation of this taxon. Our findings demonstrate that genes encoding Restriction/Modification (R/M) systems constitute a substantial part of the B. breve variable gene content (or variome). Using the methylome data generated by SMRT sequencing, combined with targeted Illumina bisulfite sequencing (BS-seq) and comparative genome analysis, we were able to detect methylation recognition motifs and assign these to identified B. breve R/M systems, where in several cases such assignments were confirmed by restriction analysis. Furthermore, we show that R/M systems typically impose a very significant barrier to genetic accessibility of B. breve strains, and that cloning of a methyltransferase-encoding gene may overcome such a barrier, thus allowing future functional investigations of members of this species.


Asunto(s)
Bifidobacterium breve/genética , Metilación de ADN , Metilasas de Modificación del ADN/genética , Genoma Bacteriano , Bifidobacterium breve/clasificación , Bifidobacterium breve/enzimología , Enzimas de Restricción del ADN/genética , Transferencia de Gen Horizontal , Genómica , Motivos de Nucleótidos , Filogenia
9.
BMC Pediatr ; 20(1): 195, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32380969

RESUMEN

BACKGROUND: It is important to understand the consequences of pre-emptive antibiotic treatment in neonates, as disturbances in microbiota development during this key developmental time window might affect early and later life health outcomes. Despite increasing knowledge regarding the detrimental effect of antibiotics on the gut microbiota, limited research focussed on antibiotic treatment duration. We determined the effect of short and long amoxicillin/ceftazidime administration on gut microbiota development during the immediate postnatal life of preterm and term infants. METHODS: Faeces was collected from 63 (pre) term infants at postnatal weeks one, two, three, four and six. Infants received either no (control), short-term (ST) or long-term (LT) postpartum amoxicillin/ceftazidime treatment. RESULTS: Compared to control infants, ST and LT infants' microbiota contained significantly higher abundance of Enterococcus during the first two postnatal weeks at the expense of Bifidobacterium and Streptococcus. Short and long antibiotic treatment both allowed for microbiota restoration within the first six postnatal weeks. However, Enterococcus and Bifidobacterium abundances were affected in fewer ST than LT infants. CONCLUSIONS: Intravenous amoxicillin/ceftazidime administration affects intestinal microbiota composition by decreasing the relative abundance of Escherichia-Shigella and Streptococcus, while increasing the relative abundance of Enterococcus and Lactobacillus species during the first two postnatal weeks. Thriving of enterococci at the expense of bifidobacteria and streptococci should be considered as aspect of the cost-benefit determination for antibiotic prescription.


Asunto(s)
Microbioma Gastrointestinal , Amoxicilina , Ceftazidima , Heces , Femenino , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , ARN Ribosómico 16S
10.
Crit Rev Microbiol ; 45(5-6): 613-648, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31859540

RESUMEN

Industrialisation has introduced several lifestyle changes and medical advancements but their impact on intestinal microbiota acquisition is often overlooked. Even though these consequential changes in the microbiota could contribute to the disease burden that accompanies industrialisation, such as obesity and atopic disease. A healthy intestinal microbiota is acquired early in life but its exact origin is not fully elucidated. The maternal microbiota is a likely source because the infant and mother intestinal microbiota share identical strains. Successfully transmitting microbes from mother to child requires microbes in the maternal donor, contact between the maternal source and the infant, and an acquiring infant recipient. Transmission can be altered by changes to any of those three transmission determinants: (1) maternal microbiota sources are shaped by the mother's genotype, diet, health status and perturbing antimicrobial exposure; (2) maternal contact is reduced through C-section and formula feeding and (3) engraftment in the infant recipient is determined by host habitat filtering, the established microbes and antibiotic disruptions. This review gives an overview of the possible maternal transmission routes, the disruptions thereof, and the missing links that should be addressed in future research to investigate the maternal transmissions that are crucial for obtaining a healthy infant microbiota.


Asunto(s)
Microbioma Gastrointestinal , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Intestinos/microbiología , Humanos , Lactante , Recién Nacido
11.
Mol Cell Proteomics ; 16(9): 1610-1620, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28684633

RESUMEN

OBJECTIVE: Development of the gastrointestinal tract and immune system can be modulated by the gut microbiota. Establishment of the intestinal microbiota, in its turn, is affected by host and environmental factors. As such, development of the gut microbiota is greatly impacted in preterm infants, who have an immature gut and are exposed to factors like hospitalization, caesarean section, antibiotics, and respiratory support. DESIGN: We analyzed fecal microbiota composition and activity of ten preterm infants (gestational age 25-30 weeks; birthweight 630-1750 g) during the first six postnatal weeks through metaproteomics (LC-MS/MS) and 16S-rRNA gene sequencing. RESULTS: A gestational-age-dependent microbial signature is observed, enabling microbiota-based differentiation between extremely preterm (25-27 weeks gestation) and very preterm (30 weeks gestation) infants. In very preterm infants, the intestinal microbiota developed toward a Bifidobacterium-dominated community and was associated with high abundance of proteins involved in carbohydrate and energy metabolism. Extremely preterm infants remained predominantly colonized by facultative anaerobes and were associated with proteins involved in membrane transport and translation. Delayed colonization by obligate anaerobes could be associated with antibiotic treatment and respiratory support. CONCLUSION: We speculate that gestational age and its associated intensity of care (e.g. antibiotics and respiratory support) affects intestinal microbiota composition and activity in preterm infants. As the gut microbiota plays a major role in development of the neonate, gestational age and its associated factors could set the stage for early and later life health complications via interference with microbiota development.


Asunto(s)
Microbioma Gastrointestinal , Recien Nacido Prematuro/metabolismo , Proteómica/métodos , Animales , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/metabolismo , Bovinos , Heces/microbiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Edad Gestacional , Humanos , Recién Nacido , Masculino , Leche/química , Oligosacáridos/análisis , Análisis de Componente Principal , Proteínas/metabolismo , ARN Ribosómico 16S/genética , Respiración
12.
J Allergy Clin Immunol ; 141(4): 1334-1342.e5, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28866384

RESUMEN

BACKGROUND: Development of the gut microbiota in infancy is important in maturation of the immune system. Deviations in colonization patterns have been associated with allergic manifestations such as eczema, but exact microbiome dysfunctions underlying allergies remain unclear. We studied the gut microbiota of 138 infants at increased risk of allergy, participating in a clinical trial investigating the effectiveness of a partially hydrolyzed protein formula supplemented with nondigestible oligosaccharides on the prevention of eczema. OBJECTIVE: The effects of interventions and breast-feeding on fecal microbiota were investigated. Additionally, we aimed to identify microbial patterns associated with the onset of eczema. METHODS: Bacterial taxonomic compositions in the first 26 weeks of life were analyzed by using 16S rRNA gene sequencing. Additionally, fecal pH and microbial metabolite levels were measured. RESULTS: Fecal microbial composition, metabolites, and pH of infants receiving partially hydrolyzed protein formula supplemented with nondigestible oligosaccharides was closer to that of breast-fed infants than that of infants receiving standard cow's milk formula. Infants with eczema by 18 months showed discordant development of bacterial genera of Enterobacteriaceae and Parabacteroides species in the first 26 weeks, as well as decreased acquisition of lactate-utilizing bacteria producing butyrate, namely Eubacterium and Anaerostipes species, supported by increased lactate and decreased butyrate levels. CONCLUSIONS: We showed that a partially hydrolyzed protein infant formula with specific prebiotics modulated the gut microbiota closer to that of breast-fed infants. Additionally, we identified a potential link between microbial activity and onset of eczema, which might reflect a suboptimal implementation of gut microbiota at specific developmental stages in infants at high risk for allergy.


Asunto(s)
Lactancia Materna , Eccema/prevención & control , Microbioma Gastrointestinal , Fórmulas Infantiles/química , Prebióticos , Método Doble Ciego , Eccema/microbiología , Heces/microbiología , Femenino , Estudios de Seguimiento , Humanos , Lactante , Recién Nacido , Masculino , Resultado del Tratamiento
13.
Int J Mol Sci ; 20(19)2019 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-31547172

RESUMEN

Postbiotics are functional bioactive compounds, generated in a matrix during fermentation, which may be used to promote health. The term postbiotics can be regarded as an umbrella term for all synonyms and related terms of these microbial fermentation components. Therefore, postbiotics can include many different constituents including metabolites, short-chain fatty acids (SCFAs), microbial cell fractions, functional proteins, extracellular polysaccharides (EPS), cell lysates, teichoic acid, peptidoglycan-derived muropeptides and pili-type structures. Postbiotics is also a rather new term in the '-biotics' field. Where consensus exists for the definitions of pre- and probiotics, this is not yet the case for postbiotics. Here we propose a working definition and review currently known postbiotic compounds, their proposed mechanisms, clinical evidence and potential applications. Research to date indicates that postbiotics can have direct immunomodulatory and clinically relevant effects and evidence can be found for the use of postbiotics in healthy individuals to improve overall health and to relief symptoms in a range of diseases such as infant colic and in adults atopic dermatitis and different causes of diarrhea.


Asunto(s)
Microbioma Gastrointestinal , Estado Nutricional , Probióticos/uso terapéutico , Adulto , Dermatitis Atópica/microbiología , Dermatitis Atópica/terapia , Diarrea/microbiología , Diarrea/terapia , Humanos , Lactante
14.
Int J Mol Sci ; 20(6)2019 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-30901843

RESUMEN

A metaproteomic analysis was conducted on the fecal microbiome of eight infants to characterize global protein and pathway expression. Although mass spectrometry-based proteomics is now a routine tool, analysis of the microbiome presents specific technical challenges, including the complexity and dynamic range of member taxa, the need for well-annotated metagenomic databases, and high inter-protein sequence redundancy and similarity. In this study, an approach was developed for assessment of biological phenotype and metabolic status, as a functional complement to DNA sequence analysis. Fecal samples were prepared and analysed by tandem mass spectrometry and a homology-based meta-clustering strategy was used to combine peptides from multiple species into representative proteins. In total, 15,250 unique peptides were sequenced and assigned to 2154 metaclusters, which were then assigned to pathways and functional groups. Differences were noted in several pathways, consistent with the dominant genera observed in different subjects. Although this study was not powered to draw conclusions from the comparisons, the results obtained demonstrate the applicability of this approach and provide the methods needed for performing semi-quantitative comparisons of human fecal microbiome composition, physiology and metabolism, as well as a more detailed assessment of microbial composition in comparison to 16S rRNA gene sequencing.


Asunto(s)
Heces/microbiología , Microbioma Gastrointestinal , Metagenómica , Proteómica , ARN Ribosómico 16S , Antibacterianos/farmacología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Lactante , Informática/métodos , Masculino , Metagenoma , Metagenómica/métodos , Proteoma , Proteómica/métodos
16.
Pediatr Res ; 83(3): 677-686, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29155807

RESUMEN

BackgroundPrebiotics and probiotics (synbiotics) can modify gut microbiota and have potential in allergy management when combined with amino-acid-based formula (AAF) for infants with cow's milk allergy (CMA).MethodsThis multicenter, double-blind, randomized controlled trial investigated the effects of an AAF-including synbiotic blend on percentages of bifidobacteria and Eubacterium rectale/Clostridium coccoides group (ER/CC) in feces from infants with suspected non-IgE-mediated CMA. Feces from age-matched healthy breastfed infants were used as reference (healthy breastfed reference (HBR)) for primary outcomes. The CMA subjects were randomized and received test or control formula for 8 weeks. Test formula was a hypoallergenic, nutritionally complete AAF including a prebiotic blend of fructo-oligosaccharides and the probiotic strain Bifidobacterium breve M-16V. Control formula was AAF without synbiotics.ResultsA total of 35 (test) and 36 (control) subjects were randomized; HBR included 51 infants. At week 8, the median percentage of bifidobacteria was higher in the test group than in the control group (35.4% vs. 9.7%, respectively; P<0.001), whereas ER/CC was lower (9.5% vs. 24.2%, respectively; P<0.001). HBR levels of bifidobacteria and ER/CC were 55% and 6.5%, respectively.ConclusionAAF including specific synbiotics, which results in levels of bifidobacteria and ER/CC approximating levels in the HBR group, improves the fecal microbiota of infants with suspected non-IgE-mediated CMA.


Asunto(s)
Aminoácidos/química , Microbioma Gastrointestinal , Fórmulas Infantiles , Hipersensibilidad a la Leche/terapia , Simbióticos , Animales , Bovinos , Clostridium , Método Doble Ciego , Eubacterium , Femenino , Humanos , Inmunoglobulina E , Lactante , Masculino , Leche , Hipersensibilidad a la Leche/inmunología , Resultado del Tratamiento
17.
Diabetes Obes Metab ; 20(6): 1408-1418, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29460474

RESUMEN

AIMS: The metabolic state of human adults is associated with their gut microbiome. The symbiosis between host and microbiome is initiated at birth, and early life microbiome perturbation can disturb health throughout life. Here, we determined how beneficial microbiome interventions in early life affect metabolic health in adulthood. METHODS: Postnatal diets were supplemented with either prebiotics (scGOS/lcFOS) or synbiotics (scGOS/lcFOS with Bifidobacterium breve M-16 V) until post-natal (PN) day 42 in a well-established rodent model for nutritional programming. Mice were subsequently challenged with a high-fat Western-style diet (WSD) for 8 weeks. Body weight and composition were monitored, as was gut microbiota composition at PN21, 42 and 98. Markers of glucose homeostasis, lipid metabolism and host transcriptomics of 6 target tissues were determined in adulthood (PN98). RESULTS: Early life synbiotics protected mice against WSD-induced excessive fat accumulation throughout life, replicable in 2 independent European animal facilities. Adult insulin sensitivity and dyslipidaemia were improved and most pronounced changes in gene expression were observed in the ileum. We observed subtle changes in faecal microbiota composition, both in early life and in adulthood, including increased abundance of Bifidobacterium. Microbiota transplantation using samples collected from synbiotics-supplemented adolescent mice at PN42 to age-matched germ-free recipients did not transfer the beneficial phenotype, indicating that synbiotics-modified microbiota at PN42 is not sufficient to transfer long-lasting protection of metabolic health status. CONCLUSION: Together, these findings show the potential and importance of timing of synbiotic interventions in early life during crucial microbiota development as a preventive measure to lower the risk of obesity and improve metabolic health throughout life.


Asunto(s)
Bifidobacterium breve , Obesidad/prevención & control , Simbióticos/administración & dosificación , Animales , Fármacos Antiobesidad/administración & dosificación , Fármacos Antiobesidad/farmacología , Glucemia/metabolismo , Constitución Corporal/fisiología , Peso Corporal/fisiología , Colesterol/metabolismo , Dieta Occidental/efectos adversos , Femenino , Microbioma Gastrointestinal/fisiología , Íleon/metabolismo , Metabolismo de los Lípidos/fisiología , Masculino , Ratones Endogámicos C57BL , Obesidad/sangre , Fenotipo , Prebióticos/administración & dosificación
18.
Eur J Clin Microbiol Infect Dis ; 37(3): 475-483, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29368074

RESUMEN

Antibiotic treatment is common practice in the neonatal ward for the prevention and treatment of sepsis, which is one of the leading causes of mortality and morbidity in preterm infants. Although the effect of antibiotic treatment on microbiota development is well recognised, little attention has been paid to treatment duration. We studied the effect of short and long intravenous antibiotic administration on intestinal microbiota development in preterm infants. Faecal samples from 15 preterm infants (35 ± 1 weeks gestation and 2871 ± 260 g birth weight) exposed to no, short (≤ 3 days) or long (≥ 5 days) treatment with amoxicillin/ceftazidime were collected during the first six postnatal weeks. Microbiota composition was determined through 16S rRNA gene sequencing and by quantitative polymerase chain reaction (qPCR). Short and long antibiotic treat ment significantly lowered the abundance of Bifidobacterium right after treatment (p = 0.027) till postnatal week three (p = 0.028). Long treatment caused Bifidobacterium abundance to remain decreased till postnatal week six (p = 0.009). Antibiotic treatment was effective against members of the Enterobacteriaceae family, but allowed Enterococcus to thrive and remain dominant for up to two weeks after antibiotic treatment discontinuation. Community richness and diversity were not affected by antibiotic treatment, but were positively associated with postnatal age (p < 0.023) and with abundance of Bifidobacterium (p = 0.003). Intravenous antibiotic administration during the first postnatal week greatly affects the infant's gastrointestinal microbiota. However, quick antibiotic treatment cessation allows for its recovery. Disturbances in microbiota development caused by short and, more extensively, by long antibiotic treatment could affect healthy development of the infant via interference with maturation of the immune system and gastrointestinal tract.


Asunto(s)
Administración Intravenosa/estadística & datos numéricos , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Recien Nacido Prematuro/fisiología , Antibacterianos/uso terapéutico , Bifidobacterium/genética , Enterobacteriaceae/genética , Enterococcus/genética , Heces/microbiología , Femenino , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiología , Humanos , Recién Nacido , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Tiempo
19.
Antonie Van Leeuwenhoek ; 111(6): 859-873, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29460206

RESUMEN

Host glycans are paramount in regulating the symbiotic relationship between humans and their gut bacteria. The constant flux of host-secreted mucin at the mucosal layer creates a steady niche for bacterial colonization. Mucin degradation by keystone species subsequently shapes the microbial community. This study investigated the transcriptional response during mucin-driven trophic interaction between the specialised mucin-degrader Akkermansia muciniphila and a butyrogenic gut commensal Anaerostipes caccae. A. muciniphila monocultures and co-cultures with non-mucolytic A. caccae from the Lachnospiraceae family were grown anaerobically in minimal media supplemented with mucin. We analysed for growth, metabolites (HPLC analysis), microbial composition (quantitative reverse transcription PCR), and transcriptional response (RNA-seq). Mucin degradation by A. muciniphila supported the growth of A. caccae and concomitant butyrate production predominantly via the acetyl-CoA pathway. Differential expression analysis (DESeq 2) showed the presence of A. caccae induced changes in the A. muciniphila transcriptional response with increased expression of mucin degradation genes and reduced expression of ribosomal genes. Two putative operons that encode for uncharacterised proteins and an efflux system, and several two-component systems were also differentially regulated. This indicated A. muciniphila changed its transcriptional regulation in response to A. caccae. This study provides insight to understand the mucin-driven microbial ecology using metatranscriptomics. Our findings show that the expression of mucolytic enzymes by A. muciniphila increases upon the presence of a community member. This could indicate its role as a keystone species that supports the microbial community in the mucosal environment by increasing the availability of mucin sugars.


Asunto(s)
Mucosa Intestinal/metabolismo , Verrucomicrobia/metabolismo , Butiratos/metabolismo , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiología , Microbiota/genética , Mucinas/metabolismo , Transcriptoma/genética
20.
Br J Nutr ; 117(2): 209-217, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28166850

RESUMEN

Human milk contains bioactive compounds that confer a protective role against gastrointestinal infections. In order to find supplements for an infant formula able to mimic these benefits of breast-feeding, two different concepts were tested. The products consisted of the following: (1) a Bifidobacterium breve- and Streptococcus thermophilus-fermented formula and (2) a combination of short-chain galacto-oligosaccharides/long-chain fructo-oligosaccharides with pectin-derived acidic oligosaccharides. A rotavirus infection suckling rat model was used to evaluate improvements in the infectious process and in the immune response of supplemented animals. Both nutritional concepts caused amelioration of the clinical symptoms, even though this was sometimes hidden by softer stool consistency in the supplemented groups. Both products also showed certain modulation of immune response, which seemed to be enhanced earlier and was accompanied by a faster resolution of the process. The viral shedding and the in vitro blocking assay suggest that these products are able to bind the viral particles, which can result in a milder infection. In conclusion, both concepts evaluated in this study showed interesting protective properties against rotavirus infection, which deserve to be investigated further.


Asunto(s)
Bacterias , Lactancia Materna , Fermentación , Gastroenteritis/prevención & control , Leche/microbiología , Oligosacáridos/uso terapéutico , Infecciones por Rotavirus/complicaciones , Animales , Animales Recién Nacidos , Bifidobacterium , Suplementos Dietéticos , Fructosa/farmacología , Fructosa/uso terapéutico , Galactosa/farmacología , Galactosa/uso terapéutico , Gastroenteritis/etiología , Gastroenteritis/virología , Humanos , Lactante , Fórmulas Infantiles , Fenómenos Fisiológicos Nutricionales del Lactante , Leche Humana/química , Oligosacáridos/farmacología , Pectinas/química , Ratas , Rotavirus , Infecciones por Rotavirus/virología , Streptococcus thermophilus , Esparcimiento de Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA