Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Gut Microbes ; 13(1): 1986666, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34705611

RESUMO

Understanding how exogenous microbes stably colonize the animal gut is essential to reveal mechanisms of action and tailor effective probiotic treatments. Bifidobacterium species are naturally enriched in the gastrointestinal tract of breast-fed infants. Human milk oligosaccharides (HMOs) are associated with this enrichment. However, direct mechanistic proof of the importance of HMOs in this colonization is lacking given milk contains additional factors that impact the gut microbiota. This study examined mice supplemented with the HMO 2'fucosyllactose (2'FL) together with a 2'FL-consuming strain, Bifidobacterium pseudocatenulatum MP80. 2'FL supplementation creates a niche for high levels of B.p. MP80 persistence, similar to Bifidobacterium levels seen in breast-fed infants. This synergism impacted gut microbiota composition, activated anti-inflammatory pathways and protected against chemically-induced colitis. These results demonstrate that bacterial-milk glycan interactions alone drive enrichment of beneficial Bifidobacterium and provide a model for tunable colonization thus facilitating insight into mechanisms of health promotion by bifidobacteriain neonates.


Assuntos
Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/metabolismo , Colite/prevenção & controle , Leite Humano/metabolismo , Oligossacarídeos/metabolismo , Animais , Aleitamento Materno , Colite/metabolismo , Colite/microbiologia , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
Metabolites ; 10(4)2020 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-32344519

RESUMO

Oligosaccharides from human or bovine milk selectively stimulate growth or metabolism of bacteria associated with the lower gastrointestinal tract of infants. Results from complex infant-type co-cultures point toward a possible synergistic effect of combining bovine milk oligosaccharides (BMO) and lactose (LAC) on enhancing the metabolism of Bifidobacterium longum subsp. longum and inhibition of Clostridium perfringens. We examine the interaction between B. longum subsp. longum and the commensal Parabacteroides distasonis, by culturing them in mono- and co-culture with different carbohydrates available. To understand the interaction between BMO and lactose on B. longum subsp. longum and test the potential postbiotic effect on C. perfringens growth and/or metabolic activity, we inoculated C. perfringens into fresh media and compared the metabolic changes to C. perfringens in cell-free supernatant from B. longum subsp. longum fermented media. In co-culture, B. longum subsp. longum benefits from P. distasonis (commensalism), especially in a lactose-rich environment. Furthermore, B. longum subsp. longum fermentation of BMO + LAC impaired C. perfringens' ability to utilize BMO as a carbon source (potential postbiotic effect).

3.
Nat Commun ; 10(1): 4406, 2019 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-31562300

RESUMO

Antimicrobial resistance is a global public health concern, and livestock play a significant role in selecting for resistance and maintaining such reservoirs. Here we study the succession of dairy cattle resistome during early life using metagenomic sequencing, as well as the relationship between resistome, gut microbiota, and diet. In our dataset, the gut of dairy calves serves as a reservoir of 329 antimicrobial resistance genes (ARGs) presumably conferring resistance to 17 classes of antibiotics, and the abundance of ARGs declines gradually during nursing. ARGs appear to co-occur with antibacterial biocide or metal resistance genes. Colostrum is a potential source of ARGs observed in calves at day 2. The dynamic changes in the resistome are likely a result of gut microbiota assembly, which is closely associated with diet transition in dairy calves. Modifications in the resistome may be possible via early-life dietary interventions to reduce overall antimicrobial resistance.


Assuntos
Ração Animal/análise , Dieta , Farmacorresistência Bacteriana Múltipla/genética , Fezes/microbiologia , Redes Reguladoras de Genes , Genes Bacterianos/genética , Animais , Animais Recém-Nascidos , Antibacterianos/farmacologia , Bovinos , Colostro/microbiologia , Indústria de Laticínios , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Esterco/microbiologia , Metagenômica/métodos , RNA Ribossômico 16S/genética , Microbiologia do Solo
4.
Elife ; 72018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30226190

RESUMO

The factors that govern assembly of the gut microbiota are insufficiently understood. Here, we test the hypothesis that inter-individual microbiota variation can arise solely from differences in the order and timing by which the gut is colonized early in life. Experiments in which mice were inoculated in sequence either with two complex seed communities or a cocktail of four bacterial strains and a seed community revealed that colonization order influenced both the outcome of community assembly and the ecological success of individual colonizers. Historical contingency and priority effects also occurred in Rag1-/- mice, suggesting that the adaptive immune system is not a major contributor to these processes. In conclusion, this study established a measurable effect of colonization history on gut microbiota assembly in a model in which host and environmental factors were strictly controlled, illuminating a potential cause for the high levels of unexplained individuality in host-associated microbial communities.


Assuntos
Bactérias/crescimento & desenvolvimento , Microbioma Gastrointestinal , Imunidade Adaptativa , Animais , Biodiversidade , Ceco/microbiologia , Contagem de Colônia Microbiana , Proteínas de Homeodomínio/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais , Fatores de Tempo
5.
mSphere ; 3(5)2018 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-30258040

RESUMO

Bifidobacterium species are important commensals capable of dominating the infant gut microbiome, in part by producing acids that suppress growth of other taxa. Bifidobacterium species are less prone to possessing antimicrobial resistance (AMR) genes (ARGs) than other taxa that may colonize infants. Given that AMR is a growing public health crisis and ARGs are present in the gut microbiome of humans from early life, this study examines the correlation between a Bifidobacterium-dominated infant gut microbiome and AMR levels, measured by a culture-independent metagenomic approach both in early life and as infants become toddlers. In general, Bifidobacterium dominance is associated with a significant reduction in AMR in a Bangladeshi cohort, both in the number of acquired AMR genes present and in the abundance of AMR genes. However, by year 2, Bangladeshi infants had no significant differences in AMR related to their early-life Bifidobacterium levels. A generalized linear model including all infants in a previously published Swedish cohort found a significant negative association between log-transformed total AMR and Bifidobacterium levels, thus confirming the relationship between Bifidobacterium levels and AMR. In both cohorts, there was no change between early-life and later-life AMR abundance in high-Bifidobacterium infants but a significant reduction in AMR abundance in low-Bifidobacterium infants. These results support the hypothesis that early Bifidobacterium dominance of the infant gut microbiome may help reduce colonization by taxa containing ARGs.IMPORTANCE Infants are vulnerable to an array of infectious diseases, and as the gut microbiome may serve as a reservoir of AMR for pathogens, reducing the levels of AMR in infants is important to infant health. This study demonstrates that high levels of Bifidobacterium are associated with reduced levels of AMR in early life and suggests that probiotic interventions to increase infant Bifidobacterium levels have the potential to reduce AMR in infants. However, this effect is not sustained at year 2 of age in Bangladeshi infants, underscoring the need for more detailed studies of the biogeography and timing of infant AMR acquisition.


Assuntos
Antibacterianos/farmacologia , Bifidobacterium/fisiologia , Farmacorresistência Bacteriana/genética , Microbioma Gastrointestinal , Bangladesh , Bifidobacterium/genética , DNA Bacteriano/genética , Fezes/microbiologia , Feminino , Humanos , Lactente , Masculino , Metagenômica , Probióticos/uso terapêutico , Análise de Regressão
6.
Curr Opin Biotechnol ; 49: 129-139, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28866242

RESUMO

Strategies aimed at modulating the gut microbiota by using live microbes range from single strains (probiotics or live biotherapeutics) to whole non-defined fecal transplants. Although often clinically efficacious, our understanding on how microbial-based strategies modulate gut microbiome composition and function is vastly incomplete. In this review, we present a framework based on ecological theory that provides mechanistic explanations for the findings obtained in studies that attempted to modulate the gut microbiota of humans and animals using live microbes. We argue that an ecological perspective grounded in theory is necessary to interpret and predict the impact of microbiome-modulating strategies and thus advance our ability to develop improved and targeted approaches with enhanced therapeutic efficiency.


Assuntos
Bactérias/metabolismo , Ecologia , Microbioma Gastrointestinal , Animais , Humanos
7.
Am J Clin Nutr ; 106(5): 1274-1286, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28877893

RESUMO

Background: Early-life colonization of the intestinal tract is a dynamic process influenced by numerous factors. The impact of probiotic-supplemented infant formula on the composition and function of the infant gut microbiota is not well defined.Objective: We sought to determine the effects of a bifidobacteria-containing formula on the healthy human intestinal microbiome during the first year of life.Design: A double-blind, randomized, placebo-controlled study of newborn infants assigned to a standard whey-based formula containing a total of 107 colony-forming units (CFU)/g of Bifidobacterium bifidum, Bifidobacterium breve, Bifidobacterium longum, B. longum subspecies infantis (intervention), or to a control formula without bifidobacteria (placebo). Breastfed controls were included. Diversity and composition of fecal microbiota were determined by 16S ribosomal RNA gene amplicon sequencing, and metabolite profiles were analyzed by ultrahigh-performance liquid chromatography-mass spectrometry over a period of 2 y.Results: Infants (n = 106) were randomly assigned to either the interventional (n = 48) or placebo (n = 49) group; 9 infants were exclusively breastfed throughout the entire intervention period of 12 mo. Infants exposed to bifidobacteria-supplemented formula showed decreased occurrence of Bacteroides and Blautia spp. associated with changes in lipids and unknown metabolites at month 1. Microbiota and metabolite profiles of intervention and placebo groups converged during the study period, and long-term colonization (24 mo) of the supplemented Bifidobacterium strains was not detected. Significant differences in microbiota and metabolites were detected between infants fed breast milk and those fed formula (P < 0.005) and between infants birthed vaginally and those birthed by cesarean delivery (P < 0.005). No significant differences were observed between infant feeding groups regarding growth, antibiotic uptake, or other health variables (P > 0.05).Conclusion: The supplementation of bifidobacteria to infant diet can modulate the occurrence of specific bacteria and metabolites during early life with no detectable long-term effects. This trial was registered at germanctr.de as DRKS00003660.


Assuntos
Bifidobacterium , Fezes/microbiologia , Microbioma Gastrointestinal , Metaboloma , Probióticos/administração & dosagem , Aleitamento Materno , Método Duplo-Cego , Ácidos Graxos Voláteis/análise , Fezes/química , Feminino , Humanos , Lactente , Fórmulas Infantis/química , Fórmulas Infantis/microbiologia , Recém-Nascido , Intestinos/microbiologia , Masculino , Leite Humano/química , Oligossacarídeos/análise , RNA Ribossômico 18S/isolamento & purificação , Análise de Sequência de DNA
8.
Cell Host Microbe ; 20(4): 515-526, 2016 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-27693307

RESUMO

Live bacteria (such as probiotics) have long been used to modulate gut microbiota and human physiology, but their colonization is mostly transient. Conceptual understanding of the ecological principles as they apply to exogenously introduced microbes in gut ecosystems is lacking. We find that, when orally administered to humans, Bifidobacterium longum AH1206 stably persists in the gut of 30% of individuals for at least 6 months without causing gastrointestinal symptoms or impacting the composition of the resident gut microbiota. AH1206 engraftment was associated with low abundance of resident B. longum and underrepresentation of specific carbohydrate utilization genes in the pre-treatment microbiome. Thus, phylogenetic limiting and resource availability are two factors that control the niche opportunity for AH1206 colonization. These findings suggest that bacterial species and functional genes absent in the gut microbiome of individual humans can be reestablished, providing opportunities for precise and personalized microbiome reconstitution.


Assuntos
Bifidobacterium longum/crescimento & desenvolvimento , Portador Sadio , Microbioma Gastrointestinal , Microbiota , Probióticos/administração & dosagem , Administração Oral , Humanos , Fatores de Tempo
9.
Arterioscler Thromb Vasc Biol ; 36(7): 1448-56, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27230129

RESUMO

OBJECTIVE: Akkermansia muciniphila (A muciniphila) is a mucin-degrading bacterium that resides in the mucus layer whose abundance inversely correlates with body weight and the development of diabetes mellitus in mice and humans. The objective of this study was to explore the regulatory effect of A muciniphila on host lipoprotein metabolism, insulin sensitivity, and hepatic metabolic inflammation. APPROACH AND RESULTS: By establishing a novel mouse model that colonized the A muciniphila in the gastrointestinal tract of the cAMP-responsive binding protein H (CREBH)-deficient mouse and in vivo chylomicron assay, we found that increased colonization of A muciniphila in the gastrointestinal tract of wild-type mice protected mice from an acute fat load-induced hyperlipidemia compared with vehicle-treated mice. A muciniphila administration also significantly ameliorated chronic hypertriglyceridemia, improved insulin sensitivity, and prevented overproduction of postprandial chylomicrons in CREBH-null mice. Mechanistic studies revealed that increased A muciniphila colonization induced expression of low-density lipoprotein receptors and apolipoprotein E in the hepatocytes of CREBH-null mice, which facilitated the uptake of intermediate-density lipoprotein via the mediation of apolipoprotein B100 and apolipoprotein E, leading to the increased clearance of triglyceride-rich lipoprotein remnants, chylomicron remnants, and intermediate-density lipoproteins, from the circulation. Treatment with A muciniphila further improved hepatic endoplasmic reticulum stress and metabolic inflammation in CREBH-null mice. CONCLUSIONS: Increased colonization of the disease-protective gut bacteria A muciniphila protected the host from acute and chronic hyperlipidemia by enhancing the low-density lipoprotein receptor expression and alleviating hepatic endoplasmic reticulum stress and the inflammatory response in CREBH-null mice.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/deficiência , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Hipertrigliceridemia/prevenção & controle , Receptores de LDL/metabolismo , Transdução de Sinais , Triglicerídeos/sangue , Verrucomicrobia/fisiologia , Animais , Apolipoproteína B-100/metabolismo , Apolipoproteínas E/metabolismo , Biomarcadores/sangue , Quilomícrons/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Modelos Animais de Doenças , Regulação para Baixo , Estresse do Retículo Endoplasmático , Predisposição Genética para Doença , Interações Hospedeiro-Patógeno , Hipertrigliceridemia/sangue , Hipertrigliceridemia/genética , Hipertrigliceridemia/microbiologia , Resistência à Insulina , Lipoproteínas IDL/metabolismo , Fígado/metabolismo , Fígado/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Fatores de Tempo
10.
Curr Opin Gastroenterol ; 32(2): 110-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26825589

RESUMO

PURPOSE OF REVIEW: A wide range of dietary carbohydrates, including prebiotic food ingredients, fermentable fibers, and milk oligosaccharides, are able to produce significant changes in the intestinal microbiota. These shifts in the microbial community are often characterized by increased levels of bifidobacteria and lactobacilli. More recent studies have revealed that species of Faecalibacterium, Akkermansia, and other less well studied members may also be enriched. We review the implications of these recent studies on future design of prebiotics and synbiotics to promote gastrointestinal health. RECENT FINDINGS: Investigations assessing the clinical outcomes associated with dietary modification of the gut microbiota have shown systemic as well as specific health benefits. Both prebiotic oligosaccharides comprised of a linear arrangement of simple sugars, as well as fiber-rich foods containing complex carbohydrates, have been used in these trials. However, individual variability and nonresponding study participants can make the outcome of dietary interventions less predictable. In contrast, synergistic synbiotics containing prebiotics that specifically stimulate a cognate probiotic provide additional options for personalized gut therapies. SUMMARY: This review describes recent research on how prebiotics and fermentable fibers can influence the gut microbiota and result in improvements to human health.


Assuntos
Gastroenteropatias/dietoterapia , Trato Gastrointestinal/imunologia , Prebióticos , Simbióticos , Bifidobacterium , Carboidratos da Dieta/administração & dosagem , Fermentação , Gastroenteropatias/prevenção & controle , Trato Gastrointestinal/microbiologia , Promoção da Saúde , Humanos , Absorção Intestinal , Lactobacillus , Fenômenos Fisiológicos da Nutrição , Oligossacarídeos/administração & dosagem , Oligossacarídeos/metabolismo , Prebióticos/administração & dosagem , Simbióticos/administração & dosagem
11.
Cell Rep ; 11(4): 527-38, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-25892234

RESUMO

Although recent research revealed an impact of westernization on diversity and composition of the human gut microbiota, the exact consequences on metacommunity characteristics are insufficiently understood, and the underlying ecological mechanisms have not been elucidated. Here, we have compared the fecal microbiota of adults from two non-industrialized regions in Papua New Guinea (PNG) with that of United States (US) residents. Papua New Guineans harbor communities with greater bacterial diversity, lower inter-individual variation, vastly different abundance profiles, and bacterial lineages undetectable in US residents. A quantification of the ecological processes that govern community assembly identified bacterial dispersal as the dominant process that shapes the microbiome in PNG but not in the US. These findings suggest that the microbiome alterations detected in industrialized societies might arise from modern lifestyle factors limiting bacterial dispersal, which has implications for human health and the development of strategies aimed to redress the impact of westernization.


Assuntos
Microbioma Gastrointestinal/genética , Adulto , Código de Barras de DNA Taxonômico , Fezes/microbiologia , Feminino , Humanos , Masculino , Papua Nova Guiné , População Rural
12.
Appl Environ Microbiol ; 81(7): 2455-65, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25616794

RESUMO

One strategy for enhancing the establishment of probiotic bacteria in the human intestinal tract is via the parallel administration of a prebiotic, which is referred to as a synbiotic. Here we present a novel method that allows a rational selection of putative probiotic strains to be used in synbiotic applications: in vivo selection (IVS). This method consists of isolating candidate probiotic strains from fecal samples following enrichment with the respective prebiotic. To test the potential of IVS, we isolated bifidobacteria from human subjects who consumed increasing doses of galactooligosaccharides (GOS) for 9 weeks. A retrospective analysis of the fecal microbiota of one subject revealed an 8-fold enrichment in Bifidobacterium adolescentis strain IVS-1 during GOS administration. The functionality of GOS to support the establishment of IVS-1 in the gastrointestinal tract was then evaluated in rats administered the bacterial strain alone, the prebiotic alone, or the synbiotic combination. Strain-specific quantitative real-time PCR showed that the addition of GOS increased B. adolescentis IVS-1 abundance in the distal intestine by nearly 2 logs compared to rats receiving only the probiotic. Illumina 16S rRNA sequencing not only confirmed the increased establishment of IVS-1 in the intestine but also revealed that the strain was able to outcompete the resident Bifidobacterium population when provided with GOS. In conclusion, this study demonstrated that IVS can be used to successfully formulate a synergistic synbiotic that can substantially enhance the establishment and competitiveness of a putative probiotic strain in the gastrointestinal tract.


Assuntos
Bifidobacterium/efeitos dos fármacos , Bifidobacterium/isolamento & purificação , Seleção Genética , Simbióticos , Animais , DNA Bacteriano/química , DNA Bacteriano/genética , DNA Ribossômico/química , DNA Ribossômico/genética , Fezes/microbiologia , Humanos , Dados de Sequência Molecular , RNA Ribossômico 16S/genética , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de DNA
13.
J Agric Food Chem ; 62(1): 159-66, 2014 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-24359228

RESUMO

High-pressure hydrothermal treatment of cereal bran results in fragmentation of the cell wall, releasing soluble, non-digestible, feruloylated oligo- and polysaccharides (FOPS), which may be beneficial to gut health. The objectives of this study were to (1) determine treatment temperatures for production of FOPS from maize bran and wheat bran and (2) determine the fermentation properties of partially purified FOPS from maize bran and wheat bran. FOPS were produced by heating bran and water (10%, w/v) in a high-pressure stirred reactor until the slurry reached 160-200 °C (in 10 °C increments). Final temperatures of 190 °C for maize bran and 200 °C for wheat bran resulted in the highest release of FOPS (49 and 50% of starting non-starch polysaccharide, respectively). Partial purification with ion exchange and dialysis resulted in a final product containing 63 and 57% total carbohydrate and 49 and 30% FOPS, respectively (other carbohydrate was starch). Following in vitro digestion (to remove starch), in vitro fermentation revealed that wheat FOPS were more bifidogenic than maize FOPS. However, maize FOPS led to continual production of short-chain fatty acid (SCFA), resulting in the highest SCFA and butyrate production at the end of the fermentation. In addition, maize FOPS showed significantly higher antioxidant activity than wheat FOPS. This study identified a process to produce FOPS from maize bran and wheat bran and showed that, considering the overall beneficial effects, FOPS from maize bran may exhibit enhanced benefits on gut health compared to those of wheat bran.


Assuntos
Fibras na Dieta/análise , Digestão , Oligossacarídeos/química , Polissacarídeos/química , Triticum/química , Zea mays/química , Bifidobacterium/metabolismo , Fibras na Dieta/metabolismo , Fermentação , Manipulação de Alimentos , Temperatura Alta , Humanos , Modelos Biológicos , Oligossacarídeos/isolamento & purificação , Oligossacarídeos/metabolismo , Polissacarídeos/isolamento & purificação , Polissacarídeos/metabolismo , Zea mays/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...