Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Microbiologyopen ; 10(3): e1187, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34180592

RESUMO

Gut microbiota development in formula-fed and breast-fed infants is known to differ. This could relate to the usage of unmodified vegetable oil instead of mammalian fat in infant formula (IF), causing the enhanced formation of the poorly soluble soap calcium palmitate (CP) in the infant's gut. Here we investigate in vitro the possible influence of CP on the infant gut bacteria. The growth of several bacterial species dominant in the infant's gut was analyzed by culturing in media with CP. Faecalibacterium prausnitzii as a sensitive representative was analyzed in detail by scanning transmission electron microscopy, membrane staining, gas chromatography, and microbial fuel cell experiments. Of all bacteria tested, the growth of several bifidobacteria and F. prausnitzii was reduced at 0.01 mg/ml CP, Bifidobacterium infantis stopped growing completely. CP reduced the cell envelope thickness of F. prausnitzii, disturbed the cell membrane fatty acids and function of membrane proteins involved in electron transport. CP inhibited the growth of bifidobacteria and faecalibacteria. This suggests that modification of fat in IF may benefit the development of the gut microbiota in formula-fed infants by supporting the colonization of important beneficial bacteria in early life. Future clinical studies are needed to confirm this.


Assuntos
Bactérias/metabolismo , Microbioma Gastrointestinal , Ácido Palmítico/metabolismo , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Bactérias/isolamento & purificação , Faecalibacterium prausnitzii/genética , Faecalibacterium prausnitzii/crescimento & desenvolvimento , Faecalibacterium prausnitzii/metabolismo , Fezes/microbiologia , Feminino , Humanos , Lactente , Fórmulas Infantis/análise , Masculino
2.
mBio ; 12(3): e0362820, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34061597

RESUMO

ß-Mannans are hemicelluloses that are abundant in modern diets as components in seed endosperms and common additives in processed food. Currently, the collective understanding of ß-mannan saccharification in the human colon is limited to a few keystone species, which presumably liberate low-molecular-weight mannooligosaccharide fragments that become directly available to the surrounding microbial community. Here, we show that a dominant butyrate producer in the human gut, Faecalibacterium prausnitzii, is able to acquire and degrade various ß-mannooligosaccharides (ß-MOS), which are derived by the primary mannanolytic activity of neighboring gut microbiota. Detailed biochemical analyses of selected protein components from their two ß-MOS utilization loci (F. prausnitzii ß-MOS utilization loci [FpMULs]) supported a concerted model whereby the imported ß-MOS are stepwise disassembled intracellularly by highly adapted enzymes. Coculturing experiments of F. prausnitzii with the primary degraders Bacteroides ovatus and Roseburia intestinalis on polymeric ß-mannan resulted in syntrophic growth, thus confirming the high efficiency of the FpMULs' uptake system. Genomic comparison with human F. prausnitzii strains and analyses of 2,441 public human metagenomes revealed that FpMULs are highly conserved and distributed worldwide. Together, our results provide a significant advance in the knowledge of ß-mannan metabolism and the degree to which its degradation is mediated by cross-feeding interactions between prominent beneficial microbes in the human gut. IMPORTANCE Commensal butyrate-producing bacteria belonging to the Firmicutes phylum are abundant in the human gut and are crucial for maintaining health. Currently, insight is lacking into how they target otherwise indigestible dietary fibers and into the trophic interactions they establish with other glycan degraders in the competitive gut environment. By combining cultivation, genomic, and detailed biochemical analyses, this work reveals the mechanism enabling F. prausnitzii, as a model Ruminococcaceae within Firmicutes, to cross-feed and access ß-mannan-derived oligosaccharides released in the gut ecosystem by the action of primary degraders. A comprehensive survey of human gut metagenomes shows that FpMULs are ubiquitous in human populations globally, highlighting the importance of microbial metabolism of ß-mannans/ß-MOS as a common dietary component. Our findings provide a mechanistic understanding of the ß-MOS utilization capability by F. prausnitzii that may be exploited to select dietary formulations specifically boosting this beneficial symbiont, and thus butyrate production, in the gut.


Assuntos
Faecalibacterium prausnitzii/genética , Faecalibacterium prausnitzii/metabolismo , Microbioma Gastrointestinal/genética , Mananas/metabolismo , Oligossacarídeos/metabolismo , Bacteroides/genética , Bacteroides/metabolismo , Clostridiales/genética , Clostridiales/metabolismo , Colo/microbiologia , Dieta , Faecalibacterium prausnitzii/crescimento & desenvolvimento , Microbioma Gastrointestinal/fisiologia , Humanos , Mananas/classificação , Metagenômica
3.
Nutrients ; 12(11)2020 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-33105554

RESUMO

Understanding the importance of the gut microbiota (GM) in non-alcoholic fatty liver disease (NAFLD) has raised the hope for therapeutic microbes. We have shown that high hepatic fat content associated with low abundance of Faecalibacterium prausnitzii in humans and, further, the administration of F. prausnitzii prevented NAFLD in mice. Here, we aimed at targeting F. prausnitzii by prebiotic xylo-oligosaccharides (XOS) to treat NAFLD. First, the effect of XOS on F. prausnitzii growth was assessed in vitro. Then, XOS was supplemented or not with high (HFD, 60% of energy from fat) or low (LFD) fat diet for 12 weeks in Wistar rats (n = 10/group). XOS increased F. prausnitzii growth, having only a minor impact on the GM composition. When supplemented with HFD, XOS ameliorated hepatic steatosis. The underlying mechanisms involved enhanced hepatic ß-oxidation and mitochondrial respiration. Nuclear magnetic resonance (1H-NMR) analysis of cecal metabolites showed that, compared to the HFD, the LFD group had a healthier cecal short-chain fatty acid profile and on the HFD, XOS reduced cecal isovalerate and tyrosine, metabolites previously linked to NAFLD. Cecal branched-chain fatty acids associated positively and butyrate negatively with hepatic triglycerides. In conclusion, XOS supplementation can ameliorate NAFLD by improving hepatic oxidative metabolism and affecting GM.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Glucuronatos/administração & dosagem , Hepatopatia Gordurosa não Alcoólica/dietoterapia , Oligossacarídeos/administração & dosagem , Prebióticos/administração & dosagem , Animais , Composição Corporal , Ceco/metabolismo , Ceco/microbiologia , Dieta com Restrição de Gorduras , Ingestão de Energia , Metabolismo Energético , Faecalibacterium prausnitzii/crescimento & desenvolvimento , Ácidos Graxos/metabolismo , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Glucose/metabolismo , Glucuronatos/metabolismo , Glucuronatos/farmacologia , Metabolismo dos Lipídeos , Fígado/metabolismo , Masculino , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/microbiologia , Oligossacarídeos/metabolismo , Oligossacarídeos/farmacologia , Oxirredução , Ratos , Ratos Wistar , Triglicerídeos/metabolismo
4.
Sci Rep ; 10(1): 8695, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32457521

RESUMO

The development of new methods capable of more realistic modeling of microbial communities necessitates that their results be quantitatively comparable with experimental findings. In this research, a new integrated agent and constraint based modeling framework abbreviated ACBM has been proposed that integrates agent-based and constraint-based modeling approaches. ACBM models the cell population in three-dimensional space to predict spatial and temporal dynamics and metabolic interactions. When used to simulate the batch growth of C. beijerinckii and two-species communities of F. prausnitzii and B. adolescent., ACBM improved on predictions made by two previous models. Furthermore, when transcriptomic data were integrated with a metabolic model of E. coli to consider intracellular constraints in the metabolism, ACBM accurately predicted growth rate, half-rate constant, and concentration of biomass, glucose, and acidic products over time. The results also show that the framework was able to predict the metabolism changes in the early stationary compared to the log phase. Finally, ACBM was implemented to estimate starved cells under heterogeneous feeding and it was concluded that a percentage of cells are always subject to starvation in a bioreactor with high volume.


Assuntos
Microbiota , Modelos Biológicos , Interface Usuário-Computador , Técnicas de Cultura Celular por Lotes , Bifidobacterium adolescentis/crescimento & desenvolvimento , Clostridium beijerinckii/crescimento & desenvolvimento , Faecalibacterium prausnitzii/crescimento & desenvolvimento
5.
JPEN J Parenter Enteral Nutr ; 42(7): 1156-1167, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29385239

RESUMO

BACKGROUND: Clostridium difficile (CD) infection (CDI) increases patient morbidity, mortality and healthcare costs. Antibiotic treatment induces gut dysbiosis and is both a major risk factor for CD colonization and treatment of CDI. Probiotics have been trialed to support commensal gut microbiota and reduce CDI. This study investigated commensal microbe Faecalibacterium prausnitzii (FP) and a prebiotic, both known to yield butyrate and be anti-inflammatory and immunomodulatory, on CD colonization and gut integrity in mice. METHODS: Mice were randomly grouped and supplemented daily with FP, prebiotic, FP + prebiotic, FP/prebiotic supernatant, or saline throughout the entire study. Following treatment with clindamycin for 3 days, mice were exposed to CD. Feces were collected at baseline, the day after antibiotic, and 1, 3, and 5 days after CD exposure and cultured for bacterial overgrowth and CD colonization. On days 1 and 5 after CD exposure, mice were randomly euthanized, and proximal colon was dissected for histological analysis and preparation of RNA for analysis of proinflammatory and anti-inflammatory cytokines. RESULTS: Although all mice exhibited bacterial overgrowth and CD colonization, bacterial burden resolved quicker in the FP + prebiotic group. This was associated with induction and resolution of innate immune responses, anion exchanger, and tight junction protein preservation in proximal colon. CD toxin virulence potential was questionable as expression of CD toxin B receptor was depleted in the FP + prebiotic group. CONCLUSION: Supplementation with anti-inflammatory butyrate-supporting commensal bacteria and prebiotic may support innate immune responses and minimize bacterial burden and negative effects during antibiotic and CD exposure.


Assuntos
Antibacterianos/efeitos adversos , Clostridioides difficile/crescimento & desenvolvimento , Infecções por Clostridium/tratamento farmacológico , Faecalibacterium prausnitzii , Microbioma Gastrointestinal , Prebióticos , Probióticos/uso terapêutico , Animais , Proteínas de Transporte de Ânions/metabolismo , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Butiratos/metabolismo , Butiratos/farmacologia , Clindamicina/efeitos adversos , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/metabolismo , Clostridioides difficile/patogenicidade , Infecções por Clostridium/imunologia , Infecções por Clostridium/metabolismo , Infecções por Clostridium/microbiologia , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/microbiologia , Citocinas/metabolismo , Modelos Animais de Doenças , Disbiose/etiologia , Faecalibacterium prausnitzii/crescimento & desenvolvimento , Faecalibacterium prausnitzii/metabolismo , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Ratos Sprague-Dawley , Receptores Imunológicos/metabolismo , Solanum tuberosum/química , Amido/farmacologia , Amido/uso terapêutico , Proteínas de Junções Íntimas/metabolismo
6.
Nutrients ; 9(12)2017 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-29231875

RESUMO

Appropriate intestinal barrier maturation during infancy largely depends on colonization with commensal bacteria. Faecalibacterium prausnitzii is an abundant obligate anaerobe that colonizes during weaning and is thought to maintain colonic health throughout life. We previously showed that F. prausnitzii induced Toll-like receptor 2 (TLR2) activation, which is linked to enhanced tight junction formation. Therefore, we hypothesized that F. prausnitzii enhances barrier integrity, an important factor in appropriate intestinal barrier maturation. In order to test metabolically active bacteria, we used a novel apical anaerobic co-culture system that allows the survival of both obligate anaerobic bacteria and oxygen-requiring intestinal epithelial cells (Caco-2). The first aim was to optimize the culture medium to enable growth and active metabolism of F. prausnitzii while maintaining the viability and barrier integrity, as measured by trans-epithelial electrical resistance (TEER), of the Caco-2 cells. This was achieved by supplementing the apical cell culture medium with bacterial culture medium. The second aim was to test the effect of F. prausnitzii on TEER across Caco-2 cell layers. Live F. prausnitzii did not improve TEER, which indicates that its benefits are not via altering tight junction integrity. The optimization of the novel dual-environment co-culturing system performed in this research will enable the investigation of new probiotics originating from indigenous beneficial bacteria.


Assuntos
Bactérias Anaeróbias/imunologia , Faecalibacterium prausnitzii/imunologia , Imunossenescência/fisiologia , Mucosa Intestinal/imunologia , Intestino Grosso/imunologia , Células CACO-2 , Técnicas de Cocultura/métodos , Faecalibacterium prausnitzii/crescimento & desenvolvimento , Humanos , Mucosa Intestinal/microbiologia , Intestino Grosso/microbiologia
7.
Best Pract Res Clin Gastroenterol ; 31(6): 643-648, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29566907

RESUMO

Faecalibacterium prausnitzii, anaerobic bacteria, is one of the main components of gut microbiota and the most important butyrate-producing bacteria in the human colon. So far, this commensal bacterium has been considered as a bioindicator of human health, once when its population is altered (decreased), inflammatory processes are favored. Several reports in the literature highlighted that the amount of Faecalibacterium prausnitzii negatively correlates to the activity of inflammatory bowel disease and colorectal cancer. Therefore, counterbalancing dysbiosis using Faecalibacterium prausnitzii as a potential active component of probiotic formulations appears to be a promising therapeutic strategy for inflammatory bowel diseases and colorectal cancer. However, once this microbial is very sensitive to oxygen, the formulation development is a great challenge. In this review, we will focus our attention on Faecalibacterium prausnitzii biology, anti-inflammatory metabolites, modulators of this bacterium population and its impact on human health.


Assuntos
Doença/etiologia , Faecalibacterium prausnitzii/patogenicidade , Microbioma Gastrointestinal/fisiologia , Faecalibacterium prausnitzii/crescimento & desenvolvimento , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA