Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Microbiol ; 22(1): 39, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35114943

RESUMEN

BACKGROUND: Yoghurt contains live bacteria that could contribute via modulation of the gut microbiota to its reported beneficial effects such as reduced body weight gain and lower incidence of type 2 diabetes. To date, the association between yoghurt consumption and the composition of the gut microbiota is underexplored. Here we used clinical variables, metabolomics, 16S rRNA and shotgun metagenomic sequencing data collected on over 1000 predominantly female UK twins to define the link between the gut microbiota and yoghurt-associated health benefits. RESULTS: According to food frequency questionnaires (FFQ), 73% of subjects consumed yoghurt. Consumers presented a healthier diet pattern (healthy eating index: beta = 2.17 ± 0.34; P = 2.72x10-10) and improved metabolic health characterised by reduced visceral fat (beta = -28.18 ± 11.71 g; P = 0.01). According to 16S rRNA gene analyses and whole shotgun metagenomic sequencing approach consistent taxonomic variations were observed with yoghurt consumption. More specifically, we identified higher abundance of species used as yoghurt starters Streptococcus thermophilus (beta = 0.41 ± 0.051; P = 6.14x10-12) and sometimes added Bifidobacterium animalis subsp. lactis (beta = 0.30 ± 0.052; P = 1.49x10-8) in the gut of yoghurt consumers. Replication in 1103 volunteers from the LifeLines-DEEP cohort confirmed the increase of S. thermophilus among yoghurt consumers. Using food records collected the day prior to faecal sampling we showed than an increase in these two yoghurt bacteria could be transient. Metabolomics analysis revealed that B. animalis subsp. lactis was associated with 13 faecal metabolites including a 3-hydroxyoctanoic acid, known to be involved in the regulation of gut inflammation. CONCLUSIONS: Yoghurt consumption is associated with reduced visceral fat mass and changes in gut microbiome including transient increase of yoghurt-contained species (i.e. S. thermophilus and B. lactis).


Asunto(s)
Bacterias/genética , Microbioma Gastrointestinal/genética , Metaboloma , Metagenoma , Probióticos/administración & dosificación , Yogur/microbiología , Anciano , Anciano de 80 o más Años , Bacterias/clasificación , Bacterias/aislamiento & purificación , Estudios de Cohortes , Heces/microbiología , Femenino , Humanos , Masculino , Metabolómica/métodos , Metagenómica/métodos , Microbiota/genética , Persona de Mediana Edad , ARN Ribosómico 16S/genética , Encuestas y Cuestionarios , Reino Unido
2.
Proc Natl Acad Sci U S A ; 114(1): 142-147, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-27980034

RESUMEN

Hosts and their microbes have established a sophisticated communication system over many millennia. Within mammalian hosts, this dynamic cross-talk is essential for maintaining intestinal homeostasis. In a genetically susceptible host, dysbiosis of the gut microbiome and dysregulated immune responses are central to the development of inflammatory bowel disease (IBD). Previous surveys of stool from the T-bet-/-Rag2-/- IBD mouse model revealed microbial features that discriminate between health and disease states. Enterobacteriaceae expansion and increased gene abundances for benzoate degradation, two-component systems, and bacterial motility proteins pointed to the potential involvement of a catecholamine-mediated bacterial signaling axis in colitis pathogenesis. Enterobacteriaceae sense and respond to microbiota-generated signals and host-derived catecholamines through the two-component quorum-sensing Escherichia coli regulators B and C (QseBC) system. On signal detection, QseC activates a cascade to induce virulence gene expression. Although a single pathogen has not been identified as a causative agent in IBD, adherent-invasive Escherichia coli (AIEC) have been implicated. Flagellar expression is necessary for the IBD-associated AIEC strain LF82 to establish colonization. Thus, we hypothesized that qseC inactivation could reduce LF82's virulence, and found that an absence of qseC leads to down-regulated flagellar expression and motility in vitro and reduced colonization in vivo. We extend these findings on the potential of QseC-based IBD therapeutics to three preclinical IBD models, wherein we observe that QseC blockade can effectively modulate colitogenic microbiotas to reduce intestinal inflammation. Collectively, our data support a role for QseC-mediated bacterial signaling in IBD pathogenesis and indicate that QseC inhibition may be a useful microbiota-targeted approach for disease management.


Asunto(s)
Colitis/patología , Colitis/terapia , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/genética , Escherichia coli/metabolismo , Percepción de Quorum/efectos de los fármacos , Animales , Catecolaminas/metabolismo , Colitis/microbiología , Flagelos/genética , Flagelos/metabolismo , Microbioma Gastrointestinal , Regulación Bacteriana de la Expresión Génica/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Sulfonamidas/farmacología , Virulencia/genética
3.
Proc Natl Acad Sci U S A ; 112(25): 7803-8, 2015 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-26056274

RESUMEN

Beneficial microbes that target molecules and pathways, such as oxidative stress, which can negatively affect both host and microbiota, may hold promise as an inflammatory bowel disease therapy. Prior work showed that a five-strain fermented milk product (FMP) improved colitis in T-bet(-/-) Rag2(-/-) mice. By varying the number of strains used in the FMP, we found that Lactococcus lactis I-1631 was sufficient to ameliorate colitis. Using comparative genomic analyses, we identified genes unique to L. lactis I-1631 involved in oxygen respiration. Respiration of oxygen results in reactive oxygen species (ROS) generation. Also, ROS are produced at high levels during intestinal inflammation and cause tissue damage. L. lactis I-1631 possesses genes encoding enzymes that detoxify ROS, such as superoxide dismutase (SodA). Thus, we hypothesized that lactococcal SodA played a role in attenuating colitis. Inactivation of the sodA gene abolished L. lactis I-1631's beneficial effect in the T-bet(-/-) Rag2(-/-) model. Similar effects were obtained in two additional colonic inflammation models, Il10(-/-) mice and dextran sulfate sodium-treated mice. Efforts to understand how a lipophobic superoxide anion (O2 (-)) can be detoxified by cytoplasmic lactoccocal SodA led to the finding that host antimicrobial-mediated lysis is a prerequisite for SodA release and SodA's extracytoplasmic O2 (-) scavenging. L. lactis I-1631 may represent a promising vehicle to deliver antioxidant, colitis-attenuating SodA to the inflamed intestinal mucosa, and host antimicrobials may play a critical role in mediating SodA's bioaccessibility.


Asunto(s)
Colitis/metabolismo , Lactococcus lactis/metabolismo , Muramidasa/metabolismo , Superóxido Dismutasa/metabolismo , Animales , Colitis/enzimología , Colitis/microbiología , Mucosa Intestinal/enzimología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Ratones , Especies Reactivas de Oxígeno/metabolismo
4.
J Biol Chem ; 291(21): 11323-36, 2016 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-27022026

RESUMEN

To ensure optimal cell growth and separation and to adapt to environmental parameters, bacteria have to maintain a balance between cell wall (CW) rigidity and flexibility. This can be achieved by a concerted action of peptidoglycan (PG) hydrolases and PG-synthesizing/modifying enzymes. In a search for new regulatory mechanisms responsible for the maintenance of this equilibrium in Lactococcus lactis, we isolated mutants that are resistant to the PG hydrolase lysozyme. We found that 14% of the causative mutations were mapped in the guaA gene, the product of which is involved in purine metabolism. Genetic and transcriptional analyses combined with PG structure determination of the guaA mutant enabled us to reveal the pivotal role of the pyrB gene in the regulation of CW rigidity. Our results indicate that conversion of l-aspartate (l-Asp) to N-carbamoyl-l-aspartate by PyrB may reduce the amount of l-Asp available for PG synthesis and thus cause the appearance of Asp/Asn-less stem peptides in PG. Such stem peptides do not form PG cross-bridges, resulting in a decrease in PG cross-linking and, consequently, reduced PG thickness and rigidity. We hypothesize that the concurrent utilization of l-Asp for pyrimidine and PG synthesis may be part of the regulatory scheme, ensuring CW flexibility during exponential growth and rigidity in stationary phase. The fact that l-Asp availability is dependent on nucleotide metabolism, which is tightly regulated in accordance with the growth rate, provides L. lactis cells the means to ensure optimal CW plasticity without the need to control the expression of PG synthesis genes.


Asunto(s)
Lactococcus lactis/metabolismo , Nucleótidos/metabolismo , Aspartato Carbamoiltransferasa/genética , Aspartato Carbamoiltransferasa/metabolismo , Ácido Aspártico/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Pared Celular/ultraestructura , Elasticidad , Genes Bacterianos , Lactococcus lactis/genética , Lactococcus lactis/crecimiento & desarrollo , Muramidasa/farmacología , Mutación , N-Acetil Muramoil-L-Alanina Amidasa/genética , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Peptidoglicano/química , Peptidoglicano/metabolismo
6.
Proc Natl Acad Sci U S A ; 107(42): 18132-7, 2010 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-20921388

RESUMEN

Intestinal health requires the coexistence of eukaryotic self with the gut microbiota and dysregulated host-microbial interactions can result in intestinal inflammation. Here, we show that colitis improved in T-bet(-/-)Rag2(-/-) mice that consumed a fermented milk product containing Bifidobacterium animalis subsp. lactis DN-173 010 strain. A decrease in cecal pH and alterations in short chain fatty acid profiles occurred with consumption, and there were concomitant increases in the abundance of select lactate-consuming and butyrate-producing bacteria. These metabolic shifts created a nonpermissive environment for the Enterobacteriaceae recently identified as colitogenic in a T-bet(-/-)Rag2(-/-) ulcerative colitis mouse model. In addition, 16S rRNA-based analysis of the T-bet(-/-)Rag2(-/-) fecal microbiota suggest that the structure of the endogenous gut microbiota played a key role in shaping the host response to the bacterial strains studied herein. We have identified features of the gut microbiota, at the membership and functional level, associated with response to this B. lactis-containing fermented milk product, and therefore this model provides a framework for evaluating and optimizing probiotic-based functional foods.


Asunto(s)
Bifidobacterium/fisiología , Colitis/microbiología , Enterobacteriaceae/patogenicidad , Inflamación/prevención & control , Leche , Animales , Fermentación , Ratones , Ratones Noqueados
7.
Nat Commun ; 14(1): 3310, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37339957

RESUMEN

The gut microbiome is important for human health, yet modulation requires more insight into inter-individual variation. Here, we explored latent structures of the human gut microbiome across the human lifespan, applying partitioning, pseudotime, and ordination approaches to >35,000 samples. Specifically, three major gut microbiome branches were identified, within which multiple partitions were observed in adulthood, with differential abundances of species along branches. Different compositions and metabolic functions characterized the branches' tips, reflecting ecological differences. An unsupervised network analysis from longitudinal data from 745 individuals showed that partitions exhibited connected gut microbiome states rather than over-partitioning. Stability in the Bacteroides-enriched branch was associated with specific ratios of Faecalibacterium:Bacteroides. We also showed that associations with factors (intrinsic and extrinsic) could be generic, branch- or partition-specific. Our ecological framework for cross-sectional and longitudinal data allows a better understanding of overall variation in the human gut microbiome and disentangles factors associated with specific configurations.


Asunto(s)
Microbioma Gastrointestinal , Humanos , Estudios Transversales , Bacteroides/genética , ARN Ribosómico 16S/genética
8.
Gut Microbes ; 14(1): 2094664, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35916669

RESUMEN

Probiotics have been used for decades to alleviate the negative side-effects of oral antibiotics, but our mechanistic understanding on how they work is so far incomplete. Here, we performed a metagenomic analysis of the fecal microbiota in participants who underwent a 14-d Helicobacter pylori eradication therapy with or without consumption of a multi-strain probiotic intervention (L. paracasei CNCM I-1518, L. paracasei CNCM I-3689, L. rhamnosus CNCM I-3690, and four yogurt strains) in a randomized, double-blinded, controlled clinical trial. Using a strain-level analysis for detection and metagenomic determination of replication rate, ingested strains were detected and replicated transiently in fecal samples and in the gut during and following antibiotic administration. Consumption of the fermented milk product led to a significant, although modest, improvement in the recovery of microbiota composition. Stratification of participants into two groups based on the degree to which their microbiome recovered showed i) a higher fecal abundance of the probiotic L. paracasei and L. rhamnosus strains and ii) an elevated replication rate of one strain (L. paracasei CNCMI-1518) in the recovery group. Collectively, our findings show a small but measurable benefit of a fermented milk product on microbiome recovery after antibiotics, which was linked to the detection and replication of specific probiotic strains. Such functional insight can form the basis for the development of probiotic-based intervention aimed to protect gut microbiome from drug treatments.


Asunto(s)
Productos Lácteos Cultivados , Microbioma Gastrointestinal , Probióticos , Antibacterianos/uso terapéutico , Heces , Humanos , Probióticos/farmacología , Probióticos/uso terapéutico
9.
Comput Struct Biotechnol J ; 20: 1632-1641, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35465165

RESUMEN

Healthy plant-based diets rich in fermentable residues may induce gas-related symptoms, possibly mediated by the gut microbiota. We previously showed that consumption of a fermented milk product (FMP) containing Bifidobacterium animalis subsp. lactis CNCM I-2494 and lactic acid bacteria improved gastrointestinal (GI) comfort in response to a flatulogenic dietary challenge in healthy individuals. To study the effects of the FMP on gut microbiota activity from those participants, we conducted a metatranscriptomic analysis of fecal samples (n = 262), which were collected during the ingestion of a habitual diet and two series of a 3-day high-residue challenge diet, before and following 28-days of FMP consumption. Most of the FMP species were detected or found enriched upon consumption of the product. FMP mitigated the effect of a flatulogenic diet on gas-related symptoms in several ways. First, FMP consumption was associated with the depletion of gas-producing bacteria and increased hydrogen to methane conversion. It also led to the upregulation of activities such as replication and downregulation of functions related to motility and chemotaxis. Furthermore, upon FMP intake, metabolic activities such as carbohydrate metabolism, attributed to B. animalis and S. thermophilus, were enriched; these activities were coincidentally found to be negatively associated with several GI symptoms. Finally, a more connected microbial ecosystem or mutualistic relationship among microbes was found in responders to the FMP intervention. Taken together, these findings suggest that consumption of the FMP improved the tolerance of a flatulogenic diet through active interactions with the resident gut microbiota.

10.
Am J Clin Nutr ; 115(2): 432-443, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-34617562

RESUMEN

BACKGROUND: Individual diet components and specific dietary regimens have been shown to impact the gut microbiome. OBJECTIVES: Here, we explored the contribution of long-term diet by searching for dietary patterns that would best associate with the gut microbiome in a population-based cohort. METHODS: Using a priori and a posteriori approaches, we constructed dietary patterns from an FFQ completed by 1800 adults in the American Gut Project. Dietary patterns were defined as groups of participants or combinations of food variables (factors) driven by criteria ranging from individual nutrients to overall diet. We associated these patterns with 16S ribosomal RNA-based gut microbiome data for a subset of 744 participants. RESULTS: Compared to individual features (e.g., fiber and protein), or to factors representing a reduced number of dietary features, 5 a posteriori dietary patterns based on food groups were best associated with gut microbiome beta diversity (P ≤ 0.0002). Two patterns followed Prudent-like diets-Plant-Based and Flexitarian-and exhibited the highest Healthy Eating Index 2010 (HEI-2010) scores. Two other patterns presented Western-like diets with a gradient in HEI-2010 scores. A fifth pattern consisted mostly of participants following an Exclusion diet (e.g., low carbohydrate). Notably, gut microbiome alpha diversity was significantly lower in the most Western pattern compared to the Flexitarian pattern (P ≤ 0.009), and the Exclusion diet pattern was associated with low relative abundance of Bifidobacterium (P ≤ 1.2 × 10-7), which was better explained by diet than health status. CONCLUSIONS: We demonstrated that global-diet a posteriori patterns were more associated with gut microbiome variations than individual dietary features among adults in the United States. These results confirm that evaluating diet as a whole is important when studying the gut microbiome. It will also facilitate the design of more personalized dietary strategies in general populations.


Asunto(s)
Dieta Saludable/estadística & datos numéricos , Dieta/métodos , Microbioma Gastrointestinal/genética , Fenómenos Fisiológicos de la Nutrición , Adulto , Encuestas sobre Dietas , Heces/microbiología , Femenino , Humanos , Masculino , ARN Ribosómico 16S/análisis , Estados Unidos
11.
Microbiology (Reading) ; 156(Pt 6): 1600-1608, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20150235

RESUMEN

Plasmid pBaSysBioII was constructed for high-throughput analysis of gene expression in Bacillus subtilis. It is an integrative plasmid with a ligation-independent cloning (LIC) site, allowing the generation of transcriptional gfpmut3 fusions with desired promoters. Integration is by a Campbell-type event and is non-mutagenic, placing the fusion at the homologous chromosomal locus. Using phoA, murAA, gapB, ptsG and cggR promoters that are responsive to phosphate availability, growth rate and carbon source, we show that detailed profiles of promoter activity can be established, with responses to changing conditions being measurable within 1 min of the stimulus. This makes pBaSysBioII a highly versatile tool for real-time gene expression analysis in growing cells of B. subtilis.


Asunto(s)
Bacillus subtilis/genética , Expresión Génica , Plásmidos , Bacillus subtilis/metabolismo , Secuencia de Bases , Carbono/metabolismo , Regulación Bacteriana de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Datos de Secuencia Molecular , Fosfatos/metabolismo , Regiones Promotoras Genéticas , Transcripción Genética
12.
mSystems ; 5(2)2020 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-32184365

RESUMEN

Lifestyle factors, such as diet, strongly influence the structure, diversity, and composition of the microbiome. While we have witnessed over the last several years a resurgence of interest in fermented foods, no study has specifically explored the effects of their consumption on gut microbiota in large cohorts. To assess whether the consumption of fermented foods is associated with a systematic signal in the gut microbiome and metabolome, we used a multi-omic approach (16S rRNA amplicon sequencing, metagenomic sequencing, and untargeted mass spectrometry) to analyze stool samples from 6,811 individuals from the American Gut Project, including 115 individuals specifically recruited for their frequency of fermented food consumption for a targeted 4-week longitudinal study. We observed subtle but statistically significant differences between consumers and nonconsumers in beta diversity as well as differential taxa between the two groups. We found that the metabolome of fermented food consumers was enriched with conjugated linoleic acid (CLA), a putatively health-promoting molecule. Cross-omic analyses between metagenomic sequencing and mass spectrometry suggest that CLA may be driven by taxa associated with fermented food consumers. Collectively, we found modest yet persistent signatures associated with fermented food consumption that appear present in multiple -omic types which motivate further investigation of how different types of fermented food impact the gut microbiome and overall health.IMPORTANCE Public interest in the effects of fermented food on the human gut microbiome is high, but limited studies have explored the association between fermented food consumption and the gut microbiome in large cohorts. Here, we used a combination of omics-based analyses to study the relationship between the microbiome and fermented food consumption in thousands of people using both cross-sectional and longitudinal data. We found that fermented food consumers have subtle differences in their gut microbiota structure, which is enriched in conjugated linoleic acid, thought to be beneficial. The results suggest that further studies of specific kinds of fermented food and their impacts on the microbiome and health will be useful.

14.
Nat Commun ; 9(1): 2802, 2018 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-30022049

RESUMEN

Dietary lipids favor the growth of the pathobiont Bilophila wadsworthia, but the relevance of this expansion in metabolic syndrome pathogenesis is poorly understood. Here, we showed that B. wadsworthia synergizes with high fat diet (HFD) to promote higher inflammation, intestinal barrier dysfunction and bile acid dysmetabolism, leading to higher glucose dysmetabolism and hepatic steatosis. Host-microbiota transcriptomics analysis reveal pathways, particularly butanoate metabolism, which may underlie the metabolic effects mediated by B. wadsworthia. Pharmacological suppression of B. wadsworthia-associated inflammation demonstrate the bacterium's intrinsic capacity to induce a negative impact on glycemic control and hepatic function. Administration of the probiotic Lactobacillus rhamnosus CNCM I-3690 limits B. wadsworthia-induced immune and metabolic impairment by limiting its expansion, reducing inflammation and reinforcing intestinal barrier. Our results suggest a new avenue for interventions against western diet-driven inflammatory and metabolic diseases.


Asunto(s)
Bilophila/patogenicidad , Infecciones por Desulfovibrionaceae/microbiología , Grasas de la Dieta/efectos adversos , Hígado Graso/microbiología , Lacticaseibacillus rhamnosus/fisiología , Síndrome Metabólico/microbiología , Probióticos/farmacología , Animales , Bilophila/crecimiento & desarrollo , Glucemia/metabolismo , Citocinas/biosíntesis , Citocinas/genética , Infecciones por Desulfovibrionaceae/etiología , Infecciones por Desulfovibrionaceae/metabolismo , Infecciones por Desulfovibrionaceae/terapia , Dieta Alta en Grasa/efectos adversos , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso/terapia , Microbioma Gastrointestinal , Hígado/microbiología , Hígado/patología , Pruebas de Función Hepática , Masculino , Redes y Vías Metabólicas/genética , Síndrome Metabólico/etiología , Síndrome Metabólico/metabolismo , Síndrome Metabólico/terapia , Ratones , Ratones Endogámicos C57BL , Transcriptoma
15.
Trends Microbiol ; 25(2): 100-112, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27916707

RESUMEN

The spread of the Western lifestyle has been accompanied by microbial changes thought to underlie the emergence of chronic, nontransmissible, immune-related diseases. The past decade has seen the unprecedented development of therapies for 'replenishing' the microbiota of sick individuals. However, functional and ecological solutions helping the host and the gut microbiota to cope with the ecological stressors of modern life are still lacking. In this review, we discuss how recent advances in gut microbiome science are leading to the identification of microbe-derived and health-relevant metabolites. These molecules will guide the selection of the next-generation of probiotics and dietary recommendations, which should also take the resident gut microbiota into account, to optimise efficacy. These solutions for maintaining a well-functioning gut ecosystem and promoting good health should be customised, palatable, and as widely accessible as possible.


Asunto(s)
Dieta , Disbiosis/microbiología , Microbioma Gastrointestinal/fisiología , Tracto Gastrointestinal/microbiología , Probióticos/análisis , Conducta Alimentaria , Humanos , Estilo de Vida , Ingesta Diaria Recomendada , Simbiosis
16.
Med Sci (Paris) ; 32(11): 999-1002, 2016 Nov.
Artículo en Francés | MEDLINE | ID: mdl-28008841

RESUMEN

Modern life is associated with changes in gut microbial communities, believed to be involved in the emergence of non-communicable chronic diseases. While there is an increasing effort of the scientific community towards designing microbiota-targeting therapies aiming to restore the microbiota of diseased patients, there is a lack of approaches designed to prevent the disruption of the symbiosis between human and its microbial symbionts. We discuss in this review how new technologies, tools and models will contribute to identify diet-derived health-relevant microbial metabolites, possible targets for dietary recommendations tailored to individuals' physiology, diet, genetics, lifestyle and gut microbiota.


Asunto(s)
Dieta , Conducta Alimentaria/fisiología , Microbioma Gastrointestinal/fisiología , Animales , Evolución Biológica , Dieta/efectos adversos , Dieta/métodos , Salud , Humanos
17.
ISME J ; 10(9): 2235-45, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26953599

RESUMEN

Resident gut microbes co-exist with transient bacteria to form the gut microbiota. Despite increasing evidence suggesting a role for transient microbes on gut microbiota function, the interplay between resident and transient members of this microbial community is poorly defined. We aimed to determine the extent to which a host's autochthonous gut microbiota influences niche permissivity to transient bacteria using a fermented milk product (FMP) as a vehicle for five food-borne bacterial strains. Using conventional and gnotobiotic rats and gut microbiome analyses (16S rRNA genes pyrosequencing and reverse transcription qPCR), we demonstrated that the clearance kinetics of one FMP bacterium, Lactococcus lactis CNCM I-1631, were dependent on the structure of the resident gut microbiota. Susceptibility of the resident gut microbiota to modulation by FMP intervention correlated with increased persistence of L. lactis. We also observed gut microbiome configurations that were associated with altered stability upon exposure to transient bacteria. Our study supports the concept that allochthonous bacteria have transient and subject-specific effects on the gut microbiome that can be leveraged to re-engineer the gut microbiome and improve dysbiosis-related diseases.


Asunto(s)
Bacterias/metabolismo , Productos Lácteos Cultivados/microbiología , Disbiosis/terapia , Microbioma Gastrointestinal , Lactococcus lactis/fisiología , Animales , Bacterias/genética , Disbiosis/microbiología , Ecología , Heces/microbiología , Vida Libre de Gérmenes , Masculino , Ratas , Análisis de Secuencia de ADN
18.
FEMS Microbiol Lett ; 242(1): 81-6, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15621423

RESUMEN

Intensity of the cholesterol-to-coprostanol conversion in the intestine, as assessed by the coprostanol-to-cholesterol ratio in faeces, was found highly variable among 15 human volunteers, ranging from absent to almost complete cholesterol conversion. The number of coprostanoligenic bacteria in the same faecal samples, as estimated by the most probable number method, was found to be less than 10(6) cellsg-1 of fresh stools in the low-to-inefficient converters and at least 10(8) cellsg-1 of fresh stools in the highest converters, indicating that the population level of cultivable faecal coprostanoligenic bacteria correlated with the intensity of cholesterol-to-coprostanol conversion in the human gut. Microbial communities of the samples were profiled by temporal temperature gradient gel electrophoresis (TTGE) of bacterial 16S rRNA gene amplicons. Dendrogram analysis of the TTGE profiles using the Pearson product moment correlation coefficient and a unweighted pair group method with arithmetic averages (UPGMA) algorithm clearly separated banding patterns from low-to-inefficient and high converters in two different clusters suggesting a relationship between TTGE profiles and coprostanoligenic activity. Principal components analysis further demonstrated that a large subset of bands rather than some individual bands contributed to this clustering.


Asunto(s)
Bacterias/metabolismo , Colestanol/metabolismo , Colesterol/metabolismo , Tracto Gastrointestinal/microbiología , Adulto , Bacterias/genética , Análisis por Conglomerados , Recuento de Colonia Microbiana , Dermatoglifia del ADN , ADN Bacteriano/análisis , ADN Bacteriano/aislamiento & purificación , ADN Ribosómico/análisis , ADN Ribosómico/aislamiento & purificación , Heces/química , Heces/microbiología , Genes de ARNr , Humanos , Persona de Mediana Edad , Filogenia , Análisis de Componente Principal , ARN Ribosómico 16S/genética
20.
Sci Rep ; 4: 6328, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-25209713

RESUMEN

The gut microbiota (GM) consists of resident commensals and transient microbes conveyed by the diet but little is known about the role of the latter on GM homeostasis. Here we show, by a conjunction of quantitative metagenomics, in silico genome reconstruction and metabolic modeling, that consumption of a fermented milk product containing dairy starters and Bifidobacterium animalis potentiates colonic short chain fatty acids production and decreases abundance of a pathobiont Bilophila wadsworthia compared to a milk product in subjects with irritable bowel syndrome (IBS, n = 28). The GM changes parallel improvement of IBS state, suggesting a role of the fermented milk bacteria in gut homeostasis. Our data challenge the view that microbes ingested with food have little impact on the human GM functioning and rather provide support for beneficial health effects.


Asunto(s)
Productos Lácteos Cultivados , Síndrome del Colon Irritable/microbiología , Microbiota/genética , Probióticos , Estómago/microbiología , Bifidobacterium/crecimiento & desarrollo , Bilophila/crecimiento & desarrollo , Butiratos/metabolismo , Dieta , Heces/microbiología , Microbiología de Alimentos , Humanos , Lactobacillus delbrueckii/crecimiento & desarrollo , Lactococcus lactis/crecimiento & desarrollo , ARN Ribosómico 16S/genética , Streptococcus thermophilus/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA