Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Commun Signal ; 22(1): 54, 2024 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-38243314

RESUMEN

BACKGROUND: The gut microbiota plays a crucial role in coronary artery disease (CAD) development, but limited attention has been given to the role of the microbiota in preventing this disease. This study aimed to identify key biomarkers using metagenomics and untargeted metabolomics and verify their associations with atherosclerosis. METHODS: A total of 371 participants, including individuals with various CAD types and CAD-free controls, were enrolled. Subsequently, significant markers were identified in the stool samples through gut metagenomic sequencing and untargeted metabolomics. In vivo and in vitro experiments were performed to investigate the mechanisms underlying the association between these markers and atherosclerosis. RESULTS: Faecal omics sequencing revealed that individuals with a substantial presence of Faecalibacterium prausnitzii had the lowest incidence of CAD across diverse CAD groups and control subjects. A random forest model confirmed the significant relationship between F. prausnitzii and CAD incidence. Notably, F. prausnitzii emerged as a robust, independent CAD predictor. Furthermore, our findings indicated the potential of the gut microbiota and gut metabolites to predict CAD occurrence and progression, potentially impacting amino acid and vitamin metabolism. F. prausnitzii mitigated inflammation and exhibited an antiatherosclerotic effect on ApoE-/- mice after gavage. This effect was attributed to reduced intestinal LPS synthesis and reinforced mechanical and mucosal barriers, leading to decreased plasma LPS levels and an antiatherosclerotic outcome. CONCLUSIONS: Sequencing of the samples revealed a previously unknown link between specific gut microbiota and atherosclerosis. Treatment with F. prausnitzii may help prevent CAD by inhibiting atherosclerosis.


Asunto(s)
Aterosclerosis , Microbioma Gastrointestinal , Humanos , Animales , Ratones , Faecalibacterium prausnitzii/metabolismo , Lipopolisacáridos
2.
Biomed Pharmacother ; 167: 115568, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37793274

RESUMEN

Ulcerative colitis (UC) is a severe inflammatory bowel disease (IBD) characterized by multifactorial complex disorders triggered by environmental factors, genetic susceptibility, and also gut microbial dysbiosis. Faecalibacterium prausnitzii, Bacteroides faecis, and Roseburia intestinalis are underrepresented species in UC patients, leading to the hypothesis that therapeutic application of those bacteria could ameliorate clinical symptoms and disease severity. Acute colitis was induced in mice by 3.5% DSS, and the commensal bacterial species were administered by oral gavage simultaneously with DSS treatment for up to 7 days. The signs of colonic inflammation, the intestinal barrier integrity, the proportion of regulatory T cells (Tregs), and the expression of pro-inflammatory and anti-inflammatory cytokines were quantified. The concentrations of SCFAs in feces were measured using Gas-liquid chromatography. The gut microbiome was analyzed in all treatment groups at the endpoint of the experiment. Results were benchmarked against a contemporary mesalazine treatment regime. We show that commensal species alone and in combination reduced disease activity index scores, inhibited colon shortening, strengthened the colonic epithelial barrier, and positively modulated tight junction protein expression. The expression level of pro-inflammatory cytokines was significantly reduced. Immune modulation occurred via inhibition of the loss of CD4 +CD25 +Treg cells in the spleen. Our study proofed that therapeutic application of F. prausnitzii, B. faecis, and R. intestinalis significantly ameliorated DSS-induced colitis at the level of clinical symptoms, histological inflammation, and immune status. Our data suggest that these positive effects are mediated by immune-modulatory pathways and influence on Treg/Th17 balance.


Asunto(s)
Colitis Ulcerosa , Colitis , Humanos , Ratones , Animales , Linfocitos T Reguladores , Faecalibacterium prausnitzii/metabolismo , Células Th17 , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/metabolismo , Colitis Ulcerosa/tratamiento farmacológico , Colon , Citocinas/metabolismo , Bacterias/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
3.
Nature ; 620(7973): 381-385, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37532933

RESUMEN

The human gut microbiota has gained interest as an environmental factor that may contribute to health or disease1. The development of next-generation probiotics is a promising strategy to modulate the gut microbiota and improve human health; however, several key candidate next-generation probiotics are strictly anaerobic2 and may require synergy with other bacteria for optimal growth. Faecalibacterium prausnitzii is a highly prevalent and abundant human gut bacterium associated with human health, but it has not yet been developed into probiotic formulations2. Here we describe the co-isolation of F. prausnitzii and Desulfovibrio piger, a sulfate-reducing bacterium, and their cross-feeding for growth and butyrate production. To produce a next-generation probiotic formulation, we adapted F. prausnitzii to tolerate oxygen exposure, and, in proof-of-concept studies, we demonstrate that the symbiotic product is tolerated by mice and humans (ClinicalTrials.gov identifier: NCT03728868 ) and is detected in the human gut in a subset of study participants. Our study describes a technology for the production of next-generation probiotics based on the adaptation of strictly anaerobic bacteria to tolerate oxygen exposures without a reduction in potential beneficial properties. Our technology may be used for the development of other strictly anaerobic strains as next-generation probiotics.


Asunto(s)
Biotecnología , Microbioma Gastrointestinal , Probióticos , Animales , Humanos , Ratones , Butiratos/metabolismo , Oxígeno/metabolismo , Oxígeno/farmacología , Probióticos/metabolismo , Aerobiosis , Faecalibacterium prausnitzii/efectos de los fármacos , Faecalibacterium prausnitzii/metabolismo , Simbiosis , Biotecnología/métodos
4.
Arthritis Res Ther ; 25(1): 130, 2023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-37496081

RESUMEN

BACKGROUND: Rheumatoid arthritis (RA) is a systemic chronic inflammatory disease that leads to joint destruction and functional disability due to the targeting of self-antigens present in the synovium, cartilage, and bone. RA is caused by a number of complex factors, including genetics, environment, dietary habits, and altered intestinal microbial flora. Microorganisms in the gut bind to nod-like receptors and Toll-like receptors to regulate the immune system and produce various metabolites, such as short-chain fatty acids (SCFAs) that interact directly with the host. Faecalibacterium prausnitzii is a representative bacterium that produces butyrate, a well-known immunomodulatory agent in the body, and this microbe exerts anti-inflammatory effects in autoimmune diseases. METHODS: In this study, F. prausnitzii was administered in a mouse model of RA, to investigate RA pathology and changes in the intestinal microbial flora. Using collagen-induced arthritic mice, which is a representative animal model of RA, we administered F. prausnitzii orally for 7 weeks. RESULTS: The arthritis score and joint tissue damage were decreased in the mice administered F. prausnitzii compared with the vehicle-treated group. In addition, administration of F. prausnitzii reduced the abundance of systemic immune cells that secrete the pro-inflammatory cytokine IL-17 and induced changes in SCFA concentrations and the intestinal microbial flora composition. It also resulted in decreased lactate and acetate concentrations, an increased butyrate concentration, and altered compositions of bacteria known to exacerbate or improve RA. CONCLUSION: These results suggest that F. prausnitzii exerts a therapeutic effect on RA by regulation of IL-17 producing cells. In addition, F. prausnitzii modify the microbial flora composition and short chain fatty acids in experimental RA mouse model.


Asunto(s)
Artritis Reumatoide , Faecalibacterium prausnitzii , Ratones , Animales , Faecalibacterium prausnitzii/metabolismo , Interleucina-17/metabolismo , Ácidos Grasos Volátiles/metabolismo , Modelos Animales de Enfermedad , Butiratos , Artritis Reumatoide/tratamiento farmacológico
5.
Artículo en Inglés | MEDLINE | ID: mdl-37277225

RESUMEN

INTRODUCTION: The commensal bacterium Faecalibacterium prausnitzii is a prominent member of the microbiome of animals and humans, and it plays an important role in several physiological processes. Numerous studies have correlated the reduction of F. prausnitzii abundance with many disease states, including irritable bowel syndrome, Crohn's disease, obesity, asthma, major depressive disorder, and metabolic diseases in humans. Studies have also correlated F. prausnitzii with diseases in humans involved in altered glucose metabolism, including diabetes. RESEARCH DESIGN AND METHODS: The aim of this study was to investigate the effects of compositions derived from three strains of F. prausnitzii (coined FPZ) on glucose metabolism in diet-induced obese male C57BL/6J prediabetic and type 2 diabetic mice. The primary endpoints of these studies were measuring changes in fasting blood glucose, glucose tolerance (as measured by a glucose tolerance test), and percent hemoglobin A1c (HbA1c) with longer term treatment. Two placebo-controlled trials were carried out using both live cell FPZ and killed cell FPZ and extracts. Two additional placebo-controlled trials were carried out in non-diabetic mice and mice that previously had type 2 diabetes (T2D). RESULTS: Both trials in prediabetic and diabetic mice revealed that peroral administration of live FPZ or extracts from FPZ lowered fasting blood glucose levels and improved glucose tolerance compared with control mice. A trial administering longer FPZ treatment also resulted in lowered percent HbA1c compared with control mice. Additionally, trials in non-diabetic mice treated with FPZ demonstrated that FPZ treatment does not lead to hypoglycemia. CONCLUSIONS: The trial results have shown that treatment with different formulations of FPZ result in lower blood glucose levels, lower percent HbA1c, and improved glucose response in mice compared with control prediabetic/diabetic mice. FPZ is a promising candidate as an orally administered probiotic or postbiotic to manage and improve pre-diabetes and T2D.


Asunto(s)
Trastorno Depresivo Mayor , Diabetes Mellitus Tipo 2 , Estado Prediabético , Humanos , Masculino , Ratones , Animales , Estado Prediabético/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Hemoglobina Glucada , Glucemia/metabolismo , Faecalibacterium prausnitzii/metabolismo , Trastorno Depresivo Mayor/tratamiento farmacológico , Ratones Endogámicos C57BL , Obesidad
6.
Br J Biomed Sci ; 80: 10794, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37025162

RESUMEN

Purpose: Additional effective therapeutic strategies for Type 2 diabetes (T2D) patients are urgently needed. Gut microbiota plays an important role in T2D development and is a promising treatment strategy for T2D patients. Faecalibacterium prausnitzii (F. prausnitzii) is regarded as one of the most important bacterial indicators for a healthy gut, but the mechanisms of its anti-diabetic properties are still unclear. Methods and Results: The abundance of F. prausnitzii in feces of patients with T2D was detected by using qPCR. The effects of F. prausnitzii on glucose homeostasis, insulin resistance (IR), dyslipidemia, hepatic steatosis and inflammation were investigated in type 2 diabetic (T2D) db/db mice. We also investigated F. prausnitzii in people. Our results showed that the abundance of F. prausnitzii was significantly lower in T2D patients compared to healthy subjects. In T2D mice, we found that F. prausnitzii treatment significantly decreased fasting blood glucose and IR index, indicating improved glucose intolerance as well as IR. Furthermore, based on evaluation of lipid-regulating enzyme activities and proinflammatory cytokine levels, F. prausnitzii was not only able to improve inflammation in both adipose tissue and liver, but also ameliorate hepatic steatosis through inhibiting the activity of hepatic lipogenic enzymes. Conclusion: These results suggested that F. prausnitzii might serve as a therapeutic option for T2D by improved IR, lipid metabolism and inflammation.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Hígado Graso , Resistencia a la Insulina , Trastornos del Metabolismo de los Lípidos , Humanos , Ratones , Animales , Faecalibacterium prausnitzii/metabolismo , Metabolismo de los Lípidos , Inflamación
7.
J Med Food ; 25(6): 565-575, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35708632

RESUMEN

Muscular atrophy is a muscle disease in which muscle mass and strength decrease due to aging, injury, metabolic disorders, or chronic conditions. Proteins in muscle tissue are degraded by the ubiquitin-proteasome pathway, and atrophy accelerates this pathway. Akkermansia muciniphila and Faecalibacterium prausnitzii strains are effective agents against metabolic and inflammatory diseases in next-generation probiotic research. In this study, we evaluated the efficacy of A. muciniphila strain EB-AMDK19 and F. prausnitzii strain EB-FPDK11 in a mouse model of muscular atrophy, since atrophy inhibits energy metabolism and immune activation. After oral administration of each strain for 4 weeks, the hind legs of the mice were fixed with a plaster cast to immobilize them for a week. As a result, the administration of EB-AMDK19 and EB-FPDK11 strains improved grip strength but did not increase muscle mass. At the molecular level, A. muciniphila and F. prausnitzii treatments decreased the expression levels of ubiquitin-proteasome genes, atrogin-1, MuRF, and cathepsin L. They increased the expression level of the mitochondrial biogenesis regulatory gene, PGC-1α. The effect of the strains was confirmed by a decrease in myostatin. Furthermore, A. muciniphila and F. prausnitzii modulated the immune function by enhancing ZO-1 and inhibiting IL-6. In particular, EB-AMDK19 promoted the expression of IL-10, an anti-inflammatory cytokine. These results suggest that A. muciniphila and F. prausnitzii may have beneficial effects on muscular atrophy, verified by newly isolated EB-AMDK19 and EB-FPDK11 as potential next-generation probiotics.


Asunto(s)
Faecalibacterium prausnitzii , Complejo de la Endopetidasa Proteasomal , Akkermansia , Animales , Faecalibacterium prausnitzii/metabolismo , Ratones , Fuerza Muscular , Atrofia Muscular/etiología , Ubiquitinas/metabolismo , Verrucomicrobia/fisiología
8.
Int J Mol Sci ; 23(3)2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-35163630

RESUMEN

The commensal bacterium Faecalibacterium prausnitzii has unique anti-inflammatory properties, at least some of which have been attributed to its production of MAM, the Microbial Anti-inflammatory Molecule. Previous phylogenetic studies of F. prausnitzii strains have revealed the existence of various phylogroups. In this work, we address the question of whether MAMs from different phylogroups display distinct anti-inflammatory properties. We first performed wide-scale identification, classification, and phylogenetic analysis of MAM-like proteins encoded in different genomes of F. prausnitzii. When combined with a gene context analysis, this approach distinguished at least 10 distinct clusters of MAMs, providing evidence for functional diversity within this protein. We then selected 11 MAMs from various clusters and evaluated their anti-inflammatory capacities in vitro. A wide range of anti-inflammatory activity was detected. MAM from the M21/2 strain had the highest inhibitory effect (96% inhibition), while MAM from reference strain A2-165 demonstrated only 56% inhibition, and MAM from strain CNCM4541 was almost inactive. These results were confirmed in vivo in murine models of acute and chronic colitis. This study provides insights into the family of MAM proteins and generates clues regarding the choice of F. prausnitzii strains as probiotics for use in targeting chronic inflammatory diseases.


Asunto(s)
Proteínas Bacterianas/genética , Faecalibacterium prausnitzii/metabolismo , Filogenia , Probióticos/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Proteínas Bacterianas/química , Proteínas Bacterianas/uso terapéutico , Secuencia de Bases , Colitis/tratamiento farmacológico , Faecalibacterium prausnitzii/genética , Variación Genética , Genoma Bacteriano , Masculino , Ratones , Análisis de Secuencia de ADN
9.
Cell Mol Life Sci ; 79(2): 76, 2022 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-35043293

RESUMEN

Probiotics currently available on the market generally belong to a narrow range of microbial species. However, recent studies about the importance of the gut microbial commensals on human health highlighted that the gut microbiome is an unexplored reservoir of potentially beneficial microbes. For this reason, academic and industrial research is focused on identifying and testing novel microbial strains of gut origin for the development of next-generation probiotics. Although several of these are promising for the prevention and treatment of many chronic diseases, studies on human subjects are still scarce and approval from regulatory agencies is, therefore, rare. In addition, some issues need to be overcome before implementing their wide application on the market, such as the best methods for cultivation and storage of these oxygen-sensitive taxa. This review summarizes the most recent evidence related to NGPs and provides an outlook to the main issues that still limit their wide employment.


Asunto(s)
Bacterias/clasificación , Bacterias/metabolismo , Microbioma Gastrointestinal/fisiología , Probióticos/farmacología , Akkermansia/metabolismo , Fenómenos Fisiológicos Bacterianos , Clostridiales/metabolismo , Disbiosis/microbiología , Faecalibacterium prausnitzii/metabolismo , Humanos , Prevotella/metabolismo
10.
Front Cell Infect Microbiol ; 12: 919701, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36683686

RESUMEN

Faecalibacterium prausnitzii is a beneficial human gut microbe and a candidate for next-generation probiotics. With probiotics now being used in clinical treatments, concerns about their safety and side effects need to be considered. Therefore, it is essential to obtain a comprehensive understanding of the genetic diversity, functional characteristics, and potential risks of different F. prausnitzii strains. In this study, we collected the genetic information of 84 F . prausnitzii strains to conduct a pan-genome analysis with multiple perspectives. Based on single-copy genes and the sequences of 16S rRNA and the compositions of the pan-genome, different phylogenetic analyses of F. prausnitzii strains were performed, which showed the genetic diversity among them. Among the proteins of the pan-genome, we found that the accessory clusters made a greater contribution to the primary genetic functions of F. prausnitzii strains than the core and specific clusters. The functional annotations of F. prausnitzii showed that only a very small number of proteins were related to human diseases and there were no secondary metabolic gene clusters encoding harmful products. At the same time, complete fatty acid metabolism was detected in F. prausnitzii. In addition, we detected harmful elements, including antibiotic resistance genes, virulence factors, and pathogenic genes, and proposed the probiotic potential risk index (PPRI) and probiotic potential risk score (PPRS) to classify these 84 strains into low-, medium-, and high-risk groups. Finally, 15 strains were identified as low-risk strains and prioritized for clinical application. Undoubtedly, our results provide a comprehensive understanding and insight into F. prausnitzii, and PPRI and PPRS can be applied to evaluate the potential risks of probiotics in general and to guide the application of probiotics in clinical application.


Asunto(s)
Faecalibacterium prausnitzii , Probióticos , Humanos , Faecalibacterium prausnitzii/genética , Faecalibacterium prausnitzii/metabolismo , Filogenia , ARN Ribosómico 16S/genética , Variación Genética
11.
PLoS Comput Biol ; 17(11): e1009060, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34723959

RESUMEN

The study of microbial communities and their interactions has attracted the interest of the scientific community, because of their potential for applications in biotechnology, ecology and medicine. The complexity of interspecies interactions, which are key for the macroscopic behavior of microbial communities, cannot be studied easily experimentally. For this reason, the modeling of microbial communities has begun to leverage the knowledge of established constraint-based methods, which have long been used for studying and analyzing the microbial metabolism of individual species based on genome-scale metabolic reconstructions of microorganisms. A main problem of genome-scale metabolic reconstructions is that they usually contain metabolic gaps due to genome misannotations and unknown enzyme functions. This problem is traditionally solved by using gap-filling algorithms that add biochemical reactions from external databases to the metabolic reconstruction, in order to restore model growth. However, gap-filling algorithms could evolve by taking into account metabolic interactions among species that coexist in microbial communities. In this work, a gap-filling method that resolves metabolic gaps at the community level was developed. The efficacy of the algorithm was tested by analyzing its ability to resolve metabolic gaps on a synthetic community of auxotrophic Escherichia coli strains. Subsequently, the algorithm was applied to resolve metabolic gaps and predict metabolic interactions in a community of Bifidobacterium adolescentis and Faecalibacterium prausnitzii, two species present in the human gut microbiota, and in an experimentally studied community of Dehalobacter and Bacteroidales species of the ACT-3 community. The community gap-filling method can facilitate the improvement of metabolic models and the identification of metabolic interactions that are difficult to identify experimentally in microbial communities.


Asunto(s)
Algoritmos , Redes y Vías Metabólicas , Microbiota/fisiología , Modelos Biológicos , Bacteroidetes/metabolismo , Bifidobacterium adolescentis/metabolismo , Biología Computacional , Simulación por Computador , Bases de Datos Factuales , Escherichia coli/metabolismo , Faecalibacterium prausnitzii/metabolismo , Microbioma Gastrointestinal/fisiología , Humanos , Peptococcaceae/metabolismo , Biología Sintética
12.
Gut Microbes ; 13(1): 1993582, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34793284

RESUMEN

Many chronic diseases are associated with decreased abundance of the gut commensal Faecalibacterium prausnitzii. This strict anaerobe can grow on dietary fibers, e.g., prebiotics, and produce high levels of butyrate, often associated to epithelial metabolism and health. However, little is known about other F. prausnitzii metabolites that may affect the colonic epithelium. Here, we analyzed prebiotic cross-feeding between F. prausnitzii and intestinal epithelial (Caco-2) cells in a "Human-oxygen Bacteria-anaerobic" coculture system. Inulin-grown F. prausnitzii enhanced Caco-2 viability and suppressed inflammation- and oxidative stress-marker expression. Inulin-grown F. prausnitzii produced excess butyrate and fructose, but only fructose efficiently promoted Caco-2 growth. Finally, fecal microbial taxonomy analysis (16S sequencing) from healthy volunteers (n = 255) showed the strongest positive correlation for F. prausnitzii abundance and stool fructose levels. We show that fructose, produced and accumulated in a fiber-rich colonic environment, supports colonic epithelium growth, while butyrate does not.


Asunto(s)
Faecalibacterium prausnitzii/metabolismo , Fructosa/metabolismo , Mucosa Intestinal/metabolismo , Inulina/metabolismo , Anaerobiosis , Butiratos/análisis , Butiratos/metabolismo , Células CACO-2 , Proliferación Celular , Supervivencia Celular , Técnicas de Cocultivo , Heces/química , Heces/microbiología , Fructosa/análisis , Microbioma Gastrointestinal , Glucosa/análisis , Glucosa/metabolismo , Transportador de Glucosa de Tipo 5/genética , Humanos , Inflamación/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/microbiología , Pectinas/metabolismo , Prebióticos
13.
Nat Commun ; 12(1): 5958, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34645820

RESUMEN

Understanding the functional potential of the gut microbiome is of primary importance for the design of innovative strategies for allergy treatment and prevention. Here we report the gut microbiome features of 90 children affected by food (FA) or respiratory (RA) allergies and 30 age-matched, healthy controls (CT). We identify specific microbial signatures in the gut microbiome of allergic children, such as higher abundance of Ruminococcus gnavus and Faecalibacterium prausnitzii, and a depletion of Bifidobacterium longum, Bacteroides dorei, B. vulgatus and fiber-degrading taxa. The metagenome of allergic children shows a pro-inflammatory potential, with an enrichment of genes involved in the production of bacterial lipo-polysaccharides and urease. We demonstrate that specific gut microbiome signatures at baseline can be predictable of immune tolerance acquisition. Finally, a strain-level selection occurring in the gut microbiome of allergic subjects is identified. R. gnavus strains enriched in FA and RA showed lower ability to degrade fiber, and genes involved in the production of a pro-inflammatory polysaccharide. We demonstrate that a gut microbiome dysbiosis occurs in allergic children, with R. gnavus emerging as a main player in pediatric allergy. These findings may open new strategies in the development of innovative preventive and therapeutic approaches. Trial: NCT04750980.


Asunto(s)
Alérgenos/inmunología , Hipersensibilidad a los Alimentos/microbiología , Microbioma Gastrointestinal/inmunología , Tolerancia Inmunológica , Hipersensibilidad Respiratoria/microbiología , Alérgenos/efectos adversos , Animales , Bacteroides/aislamiento & purificación , Bacteroides/metabolismo , Bifidobacterium longum/aislamiento & purificación , Bifidobacterium longum/metabolismo , Estudios de Casos y Controles , Niño , Preescolar , Clostridiales/aislamiento & purificación , Clostridiales/metabolismo , Alérgenos Animales/efectos adversos , Alérgenos Animales/inmunología , Huevos/efectos adversos , Faecalibacterium prausnitzii/aislamiento & purificación , Faecalibacterium prausnitzii/metabolismo , Femenino , Hipersensibilidad a los Alimentos/etiología , Hipersensibilidad a los Alimentos/inmunología , Humanos , Lipopolisacáridos/biosíntesis , Masculino , Leche/efectos adversos , Leche/inmunología , Nueces/efectos adversos , Nueces/inmunología , Polen/química , Polen/inmunología , Prunus persica/química , Prunus persica/inmunología , Pyroglyphidae/química , Pyroglyphidae/inmunología , Hipersensibilidad Respiratoria/etiología , Hipersensibilidad Respiratoria/inmunología , Ureasa/biosíntesis
14.
Mol Nutr Food Res ; 65(21): e2100433, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34558816

RESUMEN

SCOPE: Intestinal commensal microbiota interactions play critical roles in the inflammatory bowel disease (IBD) development. Candida albicans (CA) can aggravate intestinal inflammation; however, whether Faecalibacterium prausnitzii (FP) can antagonize CA is unknown. METHODS AND RESULTS: CA are co-cultured with bacteria (FP and Escherichia coli (EC)), bacterial supernatant, and bacterial medium, respectively. Then, the CA hyphae-specific genes' expression and CA cells' morphology are investigated. The Nod-like receptor pyrin-containing protein 6 (NLRP6) inflammasome, inflammatory cytokines, and antimicrobial peptides (AMPs) production are evaluated in intestinal epithelial cells pre-treated with bacteria, bacterial med, and bacterial supernatant and exposed without or with CA. Both bacteria significantly prohibit CA numbers, while only FP and FP supernatant prohibit the transformation and virulence factors (extracellular phospholipase, secreted aspartyl proteinase, and hemolysin) secretion of CA in a co-culture system compared with media controls. Further, FP and FP supernatant promote the production of the NLRP6 inflammasome, interleukin (IL)-1ß, IL-18, and antibacterial peptides (ß-defensin (BD)-2 and BD-3) and inhibit in vitro and in vivo CA growth and pathogenicity, and alleviate DSS-colitis in mice, while EC do not show the similar effect. CONCLUSION: FP improve intestinal inflammation by inhibiting CA reproduction, colonization, and pathogenicity and inducing AMP secretion in the gut. This study uncovers new relationships between intestinal microbes and fungi in IBD patients.


Asunto(s)
Colitis , Faecalibacterium prausnitzii , Animales , Candida albicans , Colitis/microbiología , Sulfato de Dextran/efectos adversos , Faecalibacterium prausnitzii/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Ratones , Virulencia
15.
Environ Microbiol ; 23(8): 4726-4740, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34296500

RESUMEN

Faecalibacterium prausnitzii is a dominant member of healthy human colon microbiota, regarded as a beneficial gut bacterium due to its ability to produce anti-inflammatory substances. However, little is known about how F. prausnitzii utilizes the nutrients present in the human gut, influencing its prevalence in the host intestinal environment. The phosphoenolpyruvate (PEP):carbohydrate phosphotransferase system (PTS) is a widely distributed and highly efficient carbohydrate transport system found in most bacterial species that catalyses the simultaneous phosphorylation and import of cognate carbohydrates; its components play physiological roles through interaction with other regulatory proteins. Here, we performed a systematic analysis of the 16 genes encoding putative PTS components (2 enzyme I, 2 HPr, and 12 enzyme II components) in F. prausnitzii A2-165. We identified the general PTS components responsible for the PEP-dependent phosphotransfer reaction and the sugar-specific PTS components involved in the transport of two carbohydrates, N-acetylglucosamine and fructose, among five enzyme II complexes. We suggest that the dissection of the functional PTS in F. prausnitzii may help to understand how this species outcompetes other bacterial species in the human intestine.


Asunto(s)
Faecalibacterium prausnitzii , Fosfotransferasas , Disección , Faecalibacterium prausnitzii/metabolismo , Humanos , Fosforilación , Fosfotransferasas/genética , Fosfotransferasas/metabolismo , Prevalencia
16.
mBio ; 12(3): e0362820, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34061597

RESUMEN

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.


Asunto(s)
Faecalibacterium prausnitzii/genética , Faecalibacterium prausnitzii/metabolismo , Microbioma Gastrointestinal/genética , Mananos/metabolismo , Oligosacáridos/metabolismo , Bacteroides/genética , Bacteroides/metabolismo , Clostridiales/genética , Clostridiales/metabolismo , Colon/microbiología , Dieta , Faecalibacterium prausnitzii/crecimiento & desarrollo , Microbioma Gastrointestinal/fisiología , Humanos , Mananos/clasificación , Metagenómica
17.
Microbiologyopen ; 10(3): e1187, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34180592

RESUMEN

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.


Asunto(s)
Bacterias/metabolismo , Microbioma Gastrointestinal , Ácido Palmítico/metabolismo , Bacterias/genética , Bacterias/crecimiento & desarrollo , Bacterias/aislamiento & purificación , Faecalibacterium prausnitzii/genética , Faecalibacterium prausnitzii/crecimiento & desarrollo , Faecalibacterium prausnitzii/metabolismo , Heces/microbiología , Femenino , Humanos , Lactante , Fórmulas Infantiles/análisis , Masculino
19.
Methods Mol Biol ; 2167: 13-24, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32712912

RESUMEN

Self-cleaving ribozymes are RNA molecules that catalyze a site-specific self-scission reaction. Analysis of self-cleavage is a crucial aspect of the biochemical study and understanding of these molecules. Here we describe a co-transcriptional assay that allows the analysis of self-cleaving ribozymes in different reaction conditions and in the presence of desired ligands and/or cofactors. Utilizing a standard T7 RNA polymerase in vitro transcription system under limiting Mg2+ concentration, followed by a 25-fold dilution of the reaction in desired conditions of self-cleavage (buffer, ions, ligands, pH, temperature, etc.) to halt the synthesis of new RNA molecules, allows the study of self-scission of these molecules without the need for purification or additional preparation steps, such as refolding procedures. Furthermore, because the transcripts are not denatured, this assay likely yields RNAs in conformations relevant to co-transcriptionally folded species in vivo.


Asunto(s)
Proteínas Bacterianas/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Pruebas de Enzimas/métodos , Faecalibacterium prausnitzii/metabolismo , Magnesio/metabolismo , ARN Catalítico/metabolismo , Transcripción Genética , Proteínas Virales/metabolismo , Proteínas Bacterianas/genética , Catálisis , Electroforesis en Gel de Poliacrilamida , Faecalibacterium prausnitzii/enzimología , Faecalibacterium prausnitzii/genética , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Iones/química , Cinética , Ligandos , Magnesio/química , Fosfoglucomutasa/metabolismo , ARN Catalítico/genética
20.
Circ Res ; 127(4): 453-465, 2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32354259

RESUMEN

RATIONALE: The elderly experience profound systemic responses after stroke, which contribute to higher mortality and more severe long-term disability. Recent studies have revealed that stroke outcomes can be influenced by the composition of gut microbiome. However, the potential benefits of manipulating the gut microbiome after injury is unknown. OBJECTIVE: To determine if restoring youthful gut microbiota after stroke aids in recovery in aged subjects, we altered the gut microbiome through young fecal transplant gavage in aged mice after experimental stroke. Further, the effect of direct enrichment of selective bacteria producing short-chain fatty acids (SCFAs) was tested as a more targeted and refined microbiome therapy. METHODS AND RESULTS: Aged male mice (18-20 months) were subjected to ischemic stroke by middle cerebral artery occlusion. We performed fecal transplant gavage 3 days after middle cerebral artery occlusion using young donor biome (2-3 months) or aged biome (18-20 months). At day 14 after stroke, aged stroke mice receiving young fecal transplant gavage had less behavioral impairment, and reduced brain and gut inflammation. Based on data from microbial sequencing and metabolomics analysis demonstrating that young fecal transplants contained much higher SCFA levels and related bacterial strains, we selected 4 SCFA-producers (Bifidobacterium longum, Clostridium symbiosum, Faecalibacterium prausnitzii, and Lactobacillus fermentum) for transplantation. These SCFA-producers alleviated poststroke neurological deficits and inflammation, and elevated gut, brain and plasma SCFA concentrations in aged stroke mice. CONCLUSIONS: This is the first study suggesting that the poor stroke recovery in aged mice can be reversed via poststroke bacteriotherapy following the replenishment of youthful gut microbiome via modulation of immunologic, microbial, and metabolomic profiles in the host.


Asunto(s)
Ácidos Grasos Volátiles/biosíntesis , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/fisiología , Infarto de la Arteria Cerebral Media/terapia , Accidente Cerebrovascular Isquémico/terapia , Factores de Edad , Animales , Bifidobacterium longum/metabolismo , Química Encefálica , Clostridium symbiosum/metabolismo , Faecalibacterium prausnitzii/metabolismo , Ácidos Grasos Volátiles/análisis , Ácidos Grasos Volátiles/sangre , Heces/química , Interleucina-17/biosíntesis , Intestinos/química , Linfocitos Intraepiteliales/fisiología , Limosilactobacillus fermentum/metabolismo , Masculino , Ratones , Mucina 2/metabolismo , Mucina 4/metabolismo , Linfocitos T Reguladores/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA