Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Appl Microbiol Biotechnol ; 107(23): 7301-7312, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37750914

RESUMO

Fluorescence-based reporter systems are valuable tools for studying gene expression dynamics in living cells. However, available strategies to follow gene expression in bacteria within their natural ecosystem that can be typically rich and complex are scarce. In this work, we designed a plasmid-based tool ensuring both the identification of a strain of interest in complex environments and the monitoring of gene expression through the combination of two distinct fluorescent proteins as reporter genes. The tool was validated in Escherichia coli to monitor the expression of eut genes involved in the catabolism of ethanolamine. We demonstrated that the constructed reporter strain gradually responds with a bimodal output to increasing ethanolamine concentrations during in vitro cultures. The reporter strain was next inoculated to mice, and flow cytometry was used to detect the reporter strain among the dense microbiota of intestinal samples and to analyze specifically the expression of eut genes. This novel dual-fluorescent reporter system would be helpful to evaluate transcriptional processes in bacteria within complex environments. KEY POINTS: • A reporter tool was developed to monitor bacterial gene expression in complex environments. • Ethanolamine utilization (eut) genes are expressed by commensal E. coli in the mouse gut. • Expression of eut genes follows a bimodal distribution.


Assuntos
Escherichia coli , Microbiota , Animais , Camundongos , Escherichia coli/genética , Escherichia coli/metabolismo , Fluorescência , Etanolamina/metabolismo , Etanolaminas , Genes Reporter , Expressão Gênica
2.
Gut ; 69(3): 487-501, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31189655

RESUMO

OBJECTIVE: To investigate whether milk polar lipids (PL) impact human intestinal lipid absorption, metabolism, microbiota and associated markers of cardiometabolic health. DESIGN: A double-blind, randomised controlled 4-week study involving 58 postmenopausal women was used to assess the chronic effects of milk PL consumption (0, 3 or 5 g-PL/day) on lipid metabolism and gut microbiota. The acute effects of milk PL on intestinal absorption and metabolism of cholesterol were assessed in a randomised controlled crossover study using tracers in ileostomy patients. RESULTS: Over 4 weeks, milk PL significantly reduced fasting and postprandial plasma concentrations of cholesterol and surrogate lipid markers of cardiovascular disease risk, including total/high-density lipoprotein-cholesterol and apolipoprotein (Apo)B/ApoA1 ratios. The highest PL dose preferentially induced a decreased number of intestine-derived chylomicron particles. Also, milk PL increased faecal loss of coprostanol, a gut-derived metabolite of cholesterol, but major bacterial populations and faecal short-chain fatty acids were not affected by milk PL, regardless of the dose. Acute ingestion of milk PL by ileostomy patients shows that milk PL decreased cholesterol absorption and increased cholesterol-ileal efflux, which can be explained by the observed co-excretion with milk sphingomyelin in the gut. CONCLUSION: The present data demonstrate for the first time in humans that milk PL can improve the cardiometabolic health by decreasing several lipid cardiovascular markers, notably through a reduced intestinal cholesterol absorption involving specific interactions in the gut, without disturbing the major bacterial phyla of gut microbiota. TRIAL REGISTRATION NUMBER: NCT02099032 and NCT02146339; Results.


Assuntos
Doenças Cardiovasculares/sangue , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipídeos/farmacologia , Sobrepeso/metabolismo , Esfingomielinas/metabolismo , Animais , Apolipoproteína A-I/sangue , Apolipoproteína B-100/sangue , Colestanol/metabolismo , Colesterol/metabolismo , HDL-Colesterol/sangue , Estudos Cross-Over , Suplementos Nutricionais , Método Duplo-Cego , Emulsificantes/farmacologia , Fezes/química , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Ileostomia , Absorção Intestinal/efeitos dos fármacos , Lipídeos/administração & dosagem , Lipídeos/análise , Pessoa de Meia-Idade , Leite/química , Pós-Menopausa , Fatores de Risco
3.
Respir Res ; 21(1): 129, 2020 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-32450847

RESUMO

BACKGROUND: While well-characterised on its molecular base, non-small cell lung cancer (NSCLC) and its interaction with local microbiota remains scarcely explored. Moreover, current studies vary in source of lung microbiota, from bronchoalveolar lavage fluid (BAL) to tissue, introducing potentially differing results. Therefore, the objective of this study was to provide detailed characterisation of the oral and multi-source lung microbiota of direct interest in lung cancer research. Since lung tumours in lower lobes (LL) have been associated with decreased survival, characteristics of the microbiota in upper (UL) and lower tumour lobes have also been examined. METHODS: Using 16S rRNA gene sequencing technology, we analysed microbiota in saliva, BAL (obtained directly on excised lobe), non-malignant, peritumoural and tumour tissue from 18 NSCLC patients eligible for surgical treatment. Detailed taxonomy, diversity and core members were provided for each microbiota, with analysis of differential abundance on all taxonomical levels (zero-inflated binomial general linear model with Benjamini-Hochberg correction), between samples and lobe locations. RESULTS: Diversity and differential abundance analysis showed clear separation of oral and lung microbiota, but more importantly, of BAL and lung tissue microbiota. Phylum Proteobacteria dominated tissue samples, while Firmicutes was more abundant in BAL and saliva (with class Clostridia and Bacilli, respectively). However, all samples showed increased abundance of phylum Firmicutes in LL, with decrease in Proteobacteria. Also, clades Actinobacteria and Flavobacteriia showed inverse abundance between BAL and extratumoural tissues depending on the lobe location. While tumour microbiota seemed the least affected by location, peritumoural tissue showed the highest susceptibility with markedly increased similarity to BAL microbiota in UL. Differences between the three lung tissues were however very limited. CONCLUSIONS: Our results confirm that BAL harbours unique lung microbiota and emphasise the importance of the sample choice for lung microbiota analysis. Further, limited differences between the tissues indicate that different local tumour-related factors, such as tumour type, stage or associated immunity, might be the ones responsible for microbiota-shaping effect. Finally, the "shift" towards Firmicutes in LL might be a sign of increased pathogenicity, as suggested in similar malignancies, and connected to worse prognosis of the LL tumours. TRIAL REGISTRATION: ClinicalTrials.gov ID: NCT03068663. Registered February 27, 2017.


Assuntos
Líquido da Lavagem Broncoalveolar/microbiologia , Carcinoma Pulmonar de Células não Pequenas/microbiologia , Neoplasias Pulmonares/microbiologia , Microbiota/fisiologia , Saliva/microbiologia , Idoso , Lavagem Broncoalveolar , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Estudos Transversais , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Saliva/metabolismo
4.
Eur J Nutr ; 57(1): 119-135, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27568059

RESUMO

PURPOSE: In the present study, we aimed to metabolically characterize the postprandial adaptations of the major tissues involved in energy, lipids and amino acids metabolisms in mini-pigs. METHOD: Mini-pigs were fed on high-fat-high-sucrose (HFHS) diet for 2 months and several tissues explored for metabolic analyses. Further, the urine metabolome was followed over the time to picture the metabolic adaptations occurring at the whole body level following overfeeding. RESULTS: After 2 months of HFHS consumption, mini-pigs displayed an obese phenotype characterized by high circulating insulin, triglycerides and cholesterol levels. At the tissue level, a general (muscle, adipose tissue, intestine) reduction in the capacity to phosphorylate glucose was observed. This was also supported by the enhanced hepatic gluconeogenesis potential, despite the concomitant normoglycaemia, suggesting that the high circulating insulin levels would be enough to maintain glucose homoeostasis. The HFHS feeding also resulted in a reduced capacity of two other pathways: the de novo lipogenesis, and the branched-chain amino acids transamination. Finally, the follow-up of the urine metabolome over the time allowed determining breaking points in the metabolic trajectory of the animals. CONCLUSIONS: Several features confirmed the pertinence of the animal model, including increased body weight, adiposity and porcine obesity index. At the metabolic level, we observed a perturbed glucose and amino acid metabolism, known to be related to the onset of the obesity. The urine metabolome analyses revealed several metabolic pathways potentially involved in the obesity onset, including TCA (citrate, pantothenic acid), amino acids catabolism (cysteine, threonine, leucine).


Assuntos
Adaptação Fisiológica/fisiologia , Dieta Hiperlipídica , Sacarose Alimentar/administração & dosagem , Porco Miniatura , Aminoácidos/metabolismo , Animais , Glicemia/metabolismo , Colesterol/sangue , Dieta Hiperlipídica/efeitos adversos , Sacarose Alimentar/efeitos adversos , Metabolismo Energético/fisiologia , Feminino , Gluconeogênese , Glucose/metabolismo , Homeostase , Hiperfagia , Insulina/sangue , Metabolismo dos Lipídeos/fisiologia , Fígado/metabolismo , Metabolômica , Fosforilação , Período Pós-Prandial/fisiologia , Suínos , Triglicerídeos/sangue , Urina/química
5.
Environ Microbiol ; 17(9): 3407-26, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25845888

RESUMO

A cellulolytic fiber-degrading bacterium, Ruminococcus champanellensis, was isolated from human faecal samples, and its genome was recently sequenced. Bioinformatic analysis of the R. champanellensis genome revealed numerous cohesin and dockerin modules, the basic elements of the cellulosome, and manual sequencing of partially sequenced genomic segments revealed two large tandem scaffoldin-coding genes that form part of a gene cluster. Representative R. champanellensis dockerins were tested against putative cohesins, and the results revealed three different cohesin-dockerin binding profiles which implied two major types of cellulosome architectures: (i) an intricate cell-bound system and (ii) a simplistic cell-free system composed of a single cohesin-containing scaffoldin. The cell-bound system can adopt various enzymatic architectures, ranging from a single enzyme to a large enzymatic complex comprising up to 11 enzymes. The variety of cellulosomal components together with adaptor proteins may infer a very tight regulation of its components. The cellulosome system of the human gut bacterium R. champanellensis closely resembles that of the bovine rumen bacterium Ruminococcus flavefaciens. The two species contain orthologous gene clusters comprising fundamental components of cellulosome architecture. Since R. champanellensis is the only human colonic bacterium known to degrade crystalline cellulose, it may thus represent a keystone species in the human gut.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Ciclo Celular/genética , Celulose/metabolismo , Celulossomas/genética , Proteínas Cromossômicas não Histona/genética , Complexos Multienzimáticos/genética , Rúmen/microbiologia , Ruminococcus/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/classificação , Sequência de Bases , Bovinos , Proteínas de Ciclo Celular/classificação , Proteínas Cromossômicas não Histona/classificação , DNA Bacteriano/genética , Fezes/microbiologia , Humanos , Dados de Sequência Molecular , Complexos Multienzimáticos/metabolismo , Família Multigênica/genética , Filogenia , Ruminococcus/genética , Ruminococcus/isolamento & purificação , Análise de Sequência de DNA , Coesinas
6.
Br J Nutr ; 114(5): 746-55, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26313935

RESUMO

Modulating the gut microbiota via dietary interventions is a common strategy to enhance the natural defence mechanisms of the host. Several in vitro studies have highlighted the probiotic potential of Bifidobacterium thermophilum RBL67 (RBL67) selected for its anti-Salmonella effects. The present study aimed to investigate the impact of RBL67 alone and combined with fructo-oligosaccharides (FOS) on the gut microbiota of Göttingen minipigs. Minipigs were fed a basal diet supplemented with 8 g/d probiotic powder (1×109 CFU/g in skim milk matrix) (probiotic diet (PRO)), 8 g/d probiotic powder plus 8 g/d FOS (synbiotic diet (SYN)) or 8 g/d skim milk powder (control), following a cross-sectional study design. Faecal and caecal microbiota compositions were analysed with pyrosequencing of 16S rRNA genes and quantitative PCR. Metabolic activity in the caecum and colon was measured by HPLC. 16S rRNA gene amplicon sequencing revealed that minipig faeces show close similarity to pig microbiota. During the treatments and at the time of killing of animals, RBL67 was consistently detected in faeces, caecum and colon at numbers of 105-106 16S rRNA copies/g content after feeding PRO and SYN diets. At the time of killing of animals, significantly higher Bifidobacterium numbers in the caecum and colon of SYN-fed minipigs were measured compared with PRO. Our data indicate that the Göttingen minipig may be a suitable model for gut microbiota research in pigs. Data from this first in vivo study of RBL67 colonisation suggest that the combination with FOS may represent a valuable symbiotic strategy to increase probiotic bacteria levels and survival in gastrointestinal tracts for feed and food applications.


Assuntos
Bifidobacterium , Intestino Grosso/microbiologia , Microbiota , Oligossacarídeos/farmacologia , Prebióticos , Probióticos , Simbióticos , Animais , Ceco/efeitos dos fármacos , Ceco/microbiologia , Colo/efeitos dos fármacos , Colo/microbiologia , Carboidratos da Dieta/farmacologia , Fezes/microbiologia , Feminino , Frutose/farmacologia , Intestino Grosso/efeitos dos fármacos , Salmonella , Suínos , Porco Miniatura
7.
Br J Nutr ; 111(12): 2135-45, 2014 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-24555487

RESUMO

The global prevalence of Fe deficiency is high and a common corrective strategy is oral Fe supplementation, which may affect the commensal gut microbiota and gastrointestinal health. The aim of the present study was to investigate the impact of different dietary Fe concentrations on the gut microbiota and gut health of rats inoculated with human faecal microbiota. Rats (8 weeks old, n 40) were divided into five (n 8 each) groups and fed diets differing only in Fe concentration during an Fe-depletion period (12 weeks) and an Fe-repletion period (4 weeks) as follows: (1) Fe-sufficient diet throughout the study period; (2) Fe-sufficient diet followed by 70 mg Fe/kg diet; (3) Fe-depleted diet throughout the study period; (4) Fe-depleted diet followed by 35 mg Fe/kg diet; (5) Fe-depleted diet followed by 70 mg Fe/kg diet. Faecal and caecal samples were analysed for gut microbiota composition (quantitative PCR and pyrosequencing) and bacterial metabolites (HPLC), and intestinal tissue samples were investigated histologically. Fe depletion did not significantly alter dominant populations of the gut microbiota and did not induce Fe-deficiency anaemia in the studied rats. Provision of the 35 mg Fe/kg diet after feeding an Fe-deficient diet significantly increased the abundance of dominant bacterial groups such as Bacteroides spp. and Clostridium cluster IV members compared with that of an Fe-deficient diet. Fe supplementation increased gut microbial butyrate concentration 6-fold compared with Fe depletion and did not affect histological colitis scores. The present results suggest that Fe supplementation enhances the concentration of beneficial gut microbiota metabolites and thus may contribute to gut health.


Assuntos
Bacteroides/crescimento & desenvolvimento , Clostridium/crescimento & desenvolvimento , Colite/prevenção & controle , Colo/microbiologia , Suplementos Nutricionais , Modelos Animais de Doenças , Ferro da Dieta/uso terapêutico , Animais , Bacteroides/imunologia , Bacteroides/isolamento & purificação , Bacteroides/metabolismo , Biomarcadores/metabolismo , Ácido Butírico/metabolismo , Ceco/crescimento & desenvolvimento , Ceco/imunologia , Ceco/metabolismo , Ceco/microbiologia , Criança , Clostridium/imunologia , Clostridium/isolamento & purificação , Clostridium/metabolismo , Colite/imunologia , Colite/metabolismo , Colite/microbiologia , Colo/crescimento & desenvolvimento , Colo/imunologia , Colo/metabolismo , Suplementos Nutricionais/efeitos adversos , Fezes/química , Fezes/microbiologia , Feminino , Vida Livre de Germes , Humanos , Íleo/crescimento & desenvolvimento , Íleo/imunologia , Íleo/metabolismo , Íleo/microbiologia , Imunidade nas Mucosas , Ferro/administração & dosagem , Ferro/análise , Deficiências de Ferro , Ferro da Dieta/efeitos adversos , Ferro da Dieta/análise , Ferro da Dieta/metabolismo , Ratos , Ratos Endogâmicos F344 , Aumento de Peso
8.
Biology (Basel) ; 13(3)2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38534413

RESUMO

The human gut microbiota plays a crucial role in maintaining host health. Our review explores the prevalence and dynamics of Enterobacteriaceae, a bacterial family within the Proteobacteria phylum, in the human gut which represents a small fraction of the gut microbiota in healthy conditions. Even though their roles are not yet fully understood, Enterobacteriaceae and especially Escherichia coli (E. coli) play a part in creating an anaerobic environment, producing vitamins and protecting against pathogenic infections. The composition and residency of E. coli strains in the gut fluctuate among individuals and is influenced by many factors such as geography, diet and health. Dysbiosis, characterized by alterations in the microbial composition of the gut microbiota, is associated with various diseases, including obesity, inflammatory bowel diseases and metabolic disorders. A consistent pattern in dysbiosis is the expansion of Proteobacteria, particularly Enterobacteriaceae, which has been proposed as a potential marker for intestinal and extra-intestinal inflammatory diseases. Here we develop the potential mechanisms contributing to Enterobacteriaceae proliferation during dysbiosis, including changes in oxygen levels, alterations in mucosal substrates and dietary factors. Better knowledge of these mechanisms is important for developing strategies to restore a balanced gut microbiota and reduce the negative consequences of the Enterobacteriaceae bloom.

9.
Res Microbiol ; 174(1-2): 103989, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35988812

RESUMO

Ethanolamine (EA) is a substrate naturally present in the human gut and its catabolism by bacteria relies on the presence of eut genes encoding specific metabolic enzymes and accessory proteins. To date, EA utilization has been mostly investigated in gut bacterial pathogens. The aim of this study was to evaluate the ability of human gut commensal Escherichia coli isolates to utilize EA as a nitrogen and/or carbon sources. Although the capacity to consume EA is heterogeneous between the 40 strains of our collection, we determined that most of them could degrade EA to generate ammonia, a useful nitrogen resource for growth. Three isolates were also able to exploit EA as a carbon source. We also revealed that the inability of some strains to catabolize EA is explained either by mutations in the eut locus or by a defect in gene transcription. Finally, we demonstrated the importance of EA utilization for an optimal fitness of commensal E. coli in vivo. Our study provides new insights on the diversity of commensal E. coli strains to utilize EA as a nutrient in the gut and opens the way for new research in the field of interactions between host, gut microbiota and pathogens.


Assuntos
Escherichia coli , Etanolamina , Humanos , Etanolamina/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Etanolaminas , Nitrogênio/metabolismo , Carbono
10.
Int J Syst Evol Microbiol ; 62(Pt 1): 138-143, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21357460

RESUMO

A strictly anaerobic, cellulolytic strain, designated 18P13(T), was isolated from a human faecal sample. Cells were Gram-positive non-motile cocci. Strain 18P13(T) was able to degrade microcrystalline cellulose but the utilization of soluble sugars was restricted to cellobiose. Acetate and succinate were the major end products of cellulose and cellobiose fermentation. 16S rRNA gene sequence analysis revealed that the isolate belonged to the genus Ruminococcus of the family Ruminococcaceae. The closest phylogenetic relative was the ruminal cellulolytic strain Ruminococcus flavefaciens ATCC 19208(T) (<95% 16S rRNA gene sequence similarity). The DNA G+C content of strain 18P13(T) was 53.05±0.7 mol%. On the basis of phylogenetic analysis, and morphological and physiological data, strain 18P13(T) can be differentiated from other members of the genus Ruminococcus with validly published names. The name Ruminococcus champanellensis sp. nov. is proposed, with 18P13(T) (=DSM 18848(T)=JCM 17042(T)) as the type strain.


Assuntos
Celulose/metabolismo , Trato Gastrointestinal/microbiologia , Ruminococcus/classificação , Ruminococcus/isolamento & purificação , Ácido Acético/metabolismo , Anaerobiose , Técnicas de Tipagem Bacteriana , Composição de Bases , Celobiose/metabolismo , Análise por Conglomerados , DNA Bacteriano/química , DNA Bacteriano/genética , DNA Ribossômico/química , DNA Ribossômico/genética , Fezes/microbiologia , Humanos , Dados de Sequência Molecular , Filogenia , RNA Ribossômico 16S/genética , Ruminococcus/genética , Ruminococcus/fisiologia , Análise de Sequência de DNA , Ácido Succínico/metabolismo
11.
Front Nutr ; 9: 902159, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36071938

RESUMO

Establishing the relationship between gut microbiota and host health has become a main target of research in the last decade. Human gut microbiota-associated animal models represent one alternative to human research, allowing for intervention studies to investigate causality. Recent cohort and in vitro studies proposed an altered gut microbiota and lactate metabolism with excessive H2 production as the main causes of infant colic. To evaluate H2 production by infant gut microbiota and to test modulation of gut colonizer lactose- and lactate-utilizer non-H2-producer, Cutibacterium avidum P279, we established and validated a gnotobiotic model using young germ-free rats inoculated with fecal slurries from infants younger than 3 months. Here, we show that infant microbiota-associated (IMA) rats inoculated with fresh feces from healthy (n = 2) and colic infants (n = 2) and fed infant formula acquired and maintained similar quantitative and qualitative fecal microbiota composition compared to the individual donor's profile. We observed that IMA rats excreted high levels of H2, which were linked to a high abundance of lactate-utilizer H2-producer Veillonella. Supplementation of C. avidum P279 to colic IMA rats reduced H2 levels compared to animals receiving a placebo. Taken together, we report high H2 production by infant gut microbiota, which might be a contributing factor for infant colic, and suggest the potential of C. avidum P279 in reducing the abdominal H2 production, bloating, and pain associated with excessive crying in colic infants.

12.
JCI Insight ; 6(10)2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-33857018

RESUMO

BACKGROUNDHigh circulating levels of ceramides (Cer) and sphingomyelins (SM) are associated with cardiometabolic diseases. The consumption of whole fat dairy products, naturally containing such polar lipids (PL), is associated with health benefits, but the impact on sphingolipidome remains unknown.METHODSIn a 4-week randomized controlled trial, 58 postmenopausal women daily consumed milk PL-enriched cream cheese (0, 3, or 5 g of milk PL). Postprandial metabolic explorations were performed before and after supplementation. Analyses included SM and Cer species in serum, chylomicrons, and feces. The ileal contents of 4 ileostomy patients were also explored after acute milk PL intake.RESULTSMilk PL decreased serum atherogenic C24:1 Cer, C16:1 SM, and C18:1 SM species (Pgroup < 0.05). Changes in serum C16+18 SM species were positively correlated with the reduction of cholesterol (r = 0.706), LDL-C (r = 0.666), and ApoB (r = 0.705) (P < 0.001). Milk PL decreased chylomicron content in total SM and C24:1 Cer (Pgroup < 0.001), parallel to a marked increase in total Cer in feces (Pgroup < 0.001). Milk PL modulated some specific SM and Cer species in both ileal efflux and feces, suggesting differential absorption and metabolization processes in the gut.CONCLUSIONMilk PL supplementation decreased atherogenic SM and Cer species associated with the improvement of cardiovascular risk markers. Our findings bring insights on sphingolipid metabolism in the gut, especially Cer, as signaling molecules potentially participating in the beneficial effects of milk PL.TRIAL REGISTRATIONClinicalTrials.gov, NCT02099032, NCT02146339.FUNDINGANR-11-ALID-007-01; PHRCI-2014: VALOBAB, no. 14-007; CNIEL; GLN 2018-11-07; HCL (sponsor).


Assuntos
Ceramidas , Metabolismo dos Lipídeos/fisiologia , Leite , Pós-Menopausa/metabolismo , Esfingomielinas , Animais , Ceramidas/análise , Ceramidas/sangue , Ceramidas/metabolismo , Queijo , Dieta , Fezes/química , Feminino , Glicolipídeos/metabolismo , Glicoproteínas/metabolismo , Humanos , Gotículas Lipídicas/metabolismo , Sobrepeso , Esfingomielinas/análise , Esfingomielinas/sangue , Esfingomielinas/metabolismo
13.
Appl Microbiol Biotechnol ; 87(6): 2097-105, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20532756

RESUMO

A xylanase gene xyn10A was isolated from the human gut bacterium Bacteroides xylanisolvens XB1A and the gene product was characterized. Xyn10A is a 40-kDa xylanase composed of a glycoside hydrolase family 10 catalytic domain with a signal peptide. A recombinant His-tagged Xyn10A was produced in Escherichia coli and purified. It was active on oat spelt and birchwood xylans and on wheat arabinoxylans. It cleaved xylotetraose, xylopentaose, and xylohexaose but not xylobiose, clearly indicating that Xyn10A is a xylanase. Surprisingly, it showed a low activity against carboxymethylcellulose but no activity at all against aryl-cellobioside and cellooligosaccharides. The enzyme exhibited K (m) and V (max) of 1.6 mg ml(-1) and 118 micromol min(-1) mg(-1) on oat spelt xylan, and its optimal temperature and pH for activity were 37 degrees C and pH 6.0, respectively. Its catalytic properties (k (cat)/K (m) = 3,300 ml mg(-1) min(-1)) suggested that Xyn10A is one of the most active GH10 xylanase described to date. Phylogenetic analyses showed that Xyn10A was closely related to other GH10 xylanases from human Bacteroides. The xyn10A gene was expressed in B. xylanisolvens XB1A cultured with glucose, xylose or xylans, and the protein was associated with the cells. Xyn10A is the first family 10 xylanase characterized from B. xylanisolvens XB1A.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Bacteroides/enzimologia , Bacteroides/isolamento & purificação , Glicosídeo Hidrolases/química , Glicosídeo Hidrolases/metabolismo , Intestinos/microbiologia , Proteínas de Bactérias/genética , Bacteroides/química , Bacteroides/classificação , Estabilidade Enzimática , Glicosídeo Hidrolases/genética , Humanos , Cinética , Dados de Sequência Molecular , Filogenia , Especificidade por Substrato
14.
Infect Immun ; 77(2): 783-90, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19064636

RESUMO

Escherichia coli O157:H7 is a food-borne pathogen causing hemorrhagic colitis and hemolytic-uremic syndrome, especially in children. The main virulence factor responsible for the more serious disease is the Shiga toxin 2 (Stx2), which is released in the gut after oral ingestion of the organism. Although it is accepted that the amount of Stx2 produced by E. coli O157:H7 in the gut is critical for the development of disease, the eukaryotic or prokaryotic gut factors that modulate Stx2 synthesis are largely unknown. In this study, we examined the influence of prokaryotic molecules released by a complex human microbiota on Stx2 synthesis by E. coli O157:H7. Stx2 synthesis was assessed after growth of E. coli O157:H7 in cecal contents of gnotobiotic rats colonized with human microbiota or in conditioned medium having supported the growth of complex human microbiota. Extracellular prokaryotic molecules produced by the commensal microbiota repress stx(2) mRNA expression and Stx2 production by inhibiting the spontaneous and induced lytic cycle mediated by RecA. These molecules, with a molecular mass of below 3 kDa, are produced in part by Bacteroides thetaiotaomicron, a predominant species of the normal human intestinal microbiota. The microbiota-induced stx(2) repression is independent of the known quorum-sensing pathways described in E. coli O157:H7 involving SdiA, QseA, QseC, or autoinducer 3. Our findings demonstrate for the first time the regulatory activity of a soluble factor produced by the complex human digestive microbiota on a bacterial virulence factor in a physiologically relevant context.


Assuntos
Bacteroides/metabolismo , Escherichia coli O157/metabolismo , Toxina Shiga II/biossíntese , Adolescente , Adulto , Animais , Ceco/microbiologia , Criança , Pré-Escolar , Regulação para Baixo , Fezes/microbiologia , Feminino , Conteúdo Gastrointestinal/microbiologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Humanos , Masculino , Percepção de Quorum , Ratos , Ratos Endogâmicos F344 , Resposta SOS em Genética , Toxina Shiga II/antagonistas & inibidores , Transcrição Gênica , Virulência , Adulto Jovem
15.
Vaccine ; 37(2): 314-324, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30503655

RESUMO

Emergence and dissemination of multidrug resistance among pathogenic Escherichia coli have posed a serious threat to public health across developing and developed countries. In combination with a flexible repertoire of virulence mechanisms, E. coli can cause a vast range of intestinal (InPEC) and extraintestinal (ExPEC) diseases but only a very limited number of antibiotics still remains effective against this pathogen. Hence, a broad spectrum E. coli vaccine could be a promising alternative to prevent the burden of such diseases, while offering the potential for covering against several InPEC and ExPEC at once. SslE, the Secreted and Surface-associated Lipoprotein of E. coli, is a widely distributed protein among InPEC and ExPEC. SslE functions ex vivo as a mucinase capable of degrading mucins and reaching the surface of mucus-producing epithelial cells. SslE was identified by reverse vaccinology as a protective vaccine candidate against an ExPEC murine model of sepsis, and further shown to be cross-effective against other ExPEC and InPEC models of infection. In this study, we aimed to gain insight into the immune response to antigen SslE and identify an immunization strategy suited to generate robust mucosal and systemic immune responses. We showed, by analyzing T cell and antibody responses, that mice immunized with SslE via an intranasal prime followed by two intramuscular boosts developed an enhanced overall immune response compared to either intranasal-only or intramuscular-only protocols. Importantly, we also report that this regimen of immunization did not impact the richness of the murine gut microbiota, and mice had a comparable cecal microbial composition, whether immunized with SslE or PBS. Collectively, our findings further support the use of SslE in future vaccination strategies to effectively target both InPEC and ExPEC while not perturbing the resident gut microbiota.


Assuntos
Infecções por Escherichia coli/prevenção & controle , Proteínas de Escherichia coli/imunologia , Vacinas contra Escherichia coli/administração & dosagem , Microbioma Gastrointestinal , Imunidade nas Mucosas , Fatores de Virulência/imunologia , Administração Intranasal , Animais , Anticorpos Antibacterianos/sangue , Citocinas/análise , Escherichia coli , Infecções por Escherichia coli/imunologia , Proteínas de Escherichia coli/administração & dosagem , Vacinas contra Escherichia coli/imunologia , Imunização Secundária , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos BALB C , Sepse/imunologia , Sepse/prevenção & controle , Fatores de Virulência/administração & dosagem
16.
FEMS Microbiol Ecol ; 66(3): 496-504, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18811647

RESUMO

The human gut harbours a wide range of bacterial communities that play key roles in supplying nutrients and energy to the host through anaerobic fermentation of dietary components and host secretions. This fermentative process involves different functional groups of microorganisms linked in a trophic chain. Although the diversity of the intestinal microbiota has been studied extensively using molecular techniques, the functional aspects of this biodiversity remain mostly unexplored. The aim of the present work was to enumerate the principal metabolic groups of microorganisms involved in the fermentative process in the gut of healthy humans. These functional groups of microorganisms were quantified by a cultural approach, while the taxonomic composition of the microbiota was assessed by in situ hybridization on the same faecal samples. The functional groups of microorganisms that predominated in the gut were the polysaccharide-degrading populations involved in the breakdown of the most readily available exogenous and endogenous substrates and the predominant butyrate-producing species. Most of the functional groups of microorganisms studied appeared to be present at rather similar levels in all healthy volunteers, suggesting that optimal numbers of these various bacterial groups are crucial for efficient gut fermentation, as well as for host nutrition and health. Significant interindividual differences were, however, confirmed with respect to the numbers of methanogenic archaea, filter paper-degrading and acetogenic bacteria and the products formed by lactate-utilizing bacteria.


Assuntos
Bactérias/classificação , Bactérias/metabolismo , Biodiversidade , Intestinos/microbiologia , Adulto , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Bactérias Anaeróbias/isolamento & purificação , Técnicas de Tipagem Bacteriana , Contagem de Colônia Microbiana , Fibras na Dieta/metabolismo , Ácidos Graxos/metabolismo , Fezes/microbiologia , Feminino , Humanos , Hidrogênio/metabolismo , Hibridização in Situ Fluorescente , Ácido Láctico/metabolismo , Masculino , Metano/metabolismo , Pessoa de Meia-Idade , Adulto Jovem
17.
Medicine (Baltimore) ; 97(50): e13676, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30558074

RESUMO

BACKGROUND: Several studies have confirmed the important role of the gut microbiota in the regulation of immune functions and its correlation with different diseases, including cancer. While brain-gut and liver-gut axes have already been demonstrated, the existence of a lung-gut axis has been suggested more recently, with the idea that changes in the gut microbiota could affect the lung microbiota, and vice versa. Likewise, the close connection between gut microbiota and cancer of proximal sites (intestines, kidneys, liver, etc.) is already well established. However, little is known whether there is a similar relation when looking at world's number one cause of death from cancer-lung cancer. OBJECTIVE: Firstly, this study aims to characterise the gut, lung, and upper airways (UAs) microbiota in patients with non-small cell lung cancer (NSCLC) treated with surgery or neoadjuvant chemotherapy plus surgery. Secondly, it aims to evaluate a chemotherapy effect on site-specific microbiota and its influence on immune profile. To our knowledge, this is the 1st study that will analyse multi-site microbiota in NSCLC patients along with site-specific immune response. METHODS: The study is a case-controlled observational trial. Forty NSCLC patients will be divided into 2 groups depending on their anamnesis: Pchir, patients eligible for surgery, or Pct-chir, patients eligible for neoadjuvant chemotherapy plus surgery. Composition of the UAs (saliva), gut (faeces), and lung microbiota (from broncho-alveolar lavage fluid (BALF) and 3 lung pieces: "healthy" tissue distal to tumour, peritumoural tissue and tumour itself) will be analysed in both groups. Immune properties will be evaluated on the local (evaluation of the tumour immune cell infiltrate, tumour classification and properties, immune cell phenotyping in BALF; human neutrophil protein (HNP) 1-3, ß-defensin 2, and calprotectin in faeces) and systemic level (blood cytokine and immune cell profile). Short-chain fatty acids (SCFAs) (major products of bacterial fermentation with an effect on immune system) will be dosed in faecal samples. Other factors such as nutrition and smoking status will be recorded for each patient. We hypothesise that smoking status and tumour type/grade will be major factors influencing both microbiota and immune/inflammatory profile of all sampling sites. Furthermore, due to non-selectivity, the same effect is expected from chemotherapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/microbiologia , Microbioma Gastrointestinal/imunologia , Neoplasias Pulmonares/microbiologia , Microbiota/imunologia , Adulto , Idoso , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/microbiologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Estudos de Casos e Controles , Fezes/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Imunidade Humoral/fisiologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/cirurgia , Microbiota/efeitos dos fármacos , Pessoa de Meia-Idade , Terapia Neoadjuvante/métodos , Saliva/imunologia , Saliva/microbiologia
18.
FEMS Microbiol Ecol ; 61(1): 121-31, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17391327

RESUMO

In humans, plant cell wall polysaccharides represent an important source of dietary fibres that are digested by gut microorganisms. Despite the extensive degradation of xylan in the colon, the population structure and the taxonomy of the predominant bacteria involved in degradation of this polysaccharide have not been extensively explored. The objective of our study was to characterize the xylanolytic microbial community from human faeces, using xylan from different botanic origins. The xylanolytic population was enumerated at high level in all faecal samples studied. The predominant xylanolytic organisms further isolated (20 strains) were assigned to Roseburia and Bacteroides species. Some Bacteroides isolates corresponded to the two newly described species Bacteroides intestinalis and Bacteroides dorei. Other isolates were closely related to Bacteroides sp. nov., a cellulolytic bacterium recently isolated from human faeces. The remaining Bacteroides strains could be considered to belong to a new species of this genus. Roseburia isolates could be assigned to the species Roseburia intestinalis. The xylanase activity of the Bacteroides and Roseburia isolates was found to be higher than that of other gut xylanolytic species previously identified. Our results provide new insights to the diversity and activity of the human gut xylanolytic community. Four new xylan-degrading Bacteroides species were identified and the xylanolytic capacity of R. intestinalis was further shown.


Assuntos
Bactérias/enzimologia , Colo/microbiologia , Fezes/microbiologia , Metano/análise , RNA Ribossômico 16S/genética , Xilanos/metabolismo , Adulto , Bactérias/genética , Bactérias/isolamento & purificação , Testes Respiratórios , Estudos de Casos e Controles , Colo/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sequência de DNA , Xilose/metabolismo , Xilosidases/metabolismo
19.
J Oncol ; 2017: 5035371, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29075294

RESUMO

The microbiota includes different microorganisms consisting of bacteria, fungi, viruses, and protozoa distributed over many human body surfaces including the skin, vagina, gut, and airways, with the highest density found in the intestine. The gut microbiota strongly influences our metabolic, endocrine, and immune systems, as well as both the peripheral and central nervous systems. Recently, a dialogue between the gut and lung microbiota has been discovered, suggesting that changes in one compartment could impact the other compartment, whether in relation to microbial composition or function. Further, this bidirectional axis is evidenced in an, either beneficial or malignant, altered immune response in one compartment following changes in the other compartment. Stimulation of the immune system arises from the microbial cells themselves, but also from their metabolites. It can be either direct or mediated by stimulated immune cells in one site impacting the other site. Additionally, this interaction may lead to immunological boost, assisting the innate immune system in its antitumour response. Thus, this review offers an insight into the composition of these sites, the gut and the lung, their role in shaping the immune system, and, finally, their role in the response to lung cancer.

20.
Artigo em Inglês | MEDLINE | ID: mdl-28484684

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) O157:H7 are human pathogens responsible for bloody diarrhea and renal failures. EHEC employ a type 3 secretion system to attach directly to the human colonic epithelium. This structure is encoded by the locus of enterocyte effacement (LEE) whose expression is regulated in response to specific nutrients. In this study, we show that the mucin-derived sugars N-acetylglucosamine (NAG) and N-acetylneuraminic acid (NANA) inhibit EHEC adhesion to epithelial cells through down-regulation of LEE expression. The effect of NAG and NANA is dependent on NagC, a transcriptional repressor of the NAG catabolism in E. coli. We show that NagC is an activator of the LEE1 operon and a critical regulator for the colonization of mice intestine by EHEC. Finally, we demonstrate that NAG and NANA as well as the metabolic activity of Bacteroides thetaiotaomicron affect the in vivo fitness of EHEC in a NagC-dependent manner. This study highlights the role of NagC in coordinating metabolism and LEE expression in EHEC and in promoting EHEC colonization in vivo.


Assuntos
Acetilglucosamina/antagonistas & inibidores , Aderência Bacteriana/efeitos dos fármacos , Escherichia coli Êntero-Hemorrágica/genética , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Fosfoproteínas/genética , Proteínas Repressoras/genética , Animais , Bacteroides thetaiotaomicron/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Escherichia coli Êntero-Hemorrágica/metabolismo , Escherichia coli Êntero-Hemorrágica/patogenicidade , Células Epiteliais/microbiologia , Escherichia coli O157/genética , Escherichia coli O157/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiologia , Células HCT116 , Células HeLa , Humanos , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Ácido N-Acetilneuramínico/antagonistas & inibidores , Óperon , Fosfoproteínas/metabolismo , Proteínas Repressoras/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA