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










Base de dados
Intervalo de ano de publicação
1.
Nat Microbiol ; 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38918470

RESUMO

Tryptophan is catabolized by gut microorganisms resulting in a wide range of metabolites implicated in both beneficial and adverse host effects. How gut microbial tryptophan metabolism is directed towards indole, associated with chronic kidney disease, or towards protective indolelactic acid (ILA) and indolepropionic acid (IPA) is unclear. Here we used in vitro culturing and animal experiments to assess gut microbial competition for tryptophan and the resulting metabolites in a controlled three-species defined community and in complex undefined human faecal communities. The generation of specific tryptophan-derived metabolites was not predominantly determined by the abundance of tryptophan-metabolizing bacteria, but rather by substrate-dependent regulation of specific metabolic pathways. Indole-producing Escherichia coli and ILA- and IPA-producing Clostridium sporogenes competed for tryptophan within the three-species community in vitro and in vivo. Importantly, fibre-degrading Bacteroides thetaiotaomicron affected this competition by cross-feeding monosaccharides to E. coli. This inhibited indole production through catabolite repression, thus making more tryptophan available to C. sporogenes, resulting in increased ILA and IPA production. The fibre-dependent reduction in indole was confirmed using human faecal cultures and faecal-microbiota-transplanted gnotobiotic mice. Our findings explain why consumption of fermentable fibres suppresses indole production but promotes the generation of other tryptophan metabolites associated with health benefits.

2.
FEMS Microbiol Lett ; 3702023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-37028942

RESUMO

The expanding knowledge of the health impacts of the metabolic activities of the gut microbiota reinforces the current interest in engineered probiotics. Tryptophan metabolites, in particular indole lactic acid (ILA), are attractive candidates as potential therapeutic agents. ILA is a promising compound with multiple beneficial effects, including amelioration colitis in rodent models of necrotizing enterocolitis, as well as improved infant immune system maturation. In this work, we engineered and characterized in vitro and in vivo an Escherichia coli Nissle 1917 strain that produces ILA. The 2-step metabolic pathway comprises aminotransferases native of E. coli and a dehydrogenase introduced from Bifidobacterium longum subspecies infantis. Our results show a robust engineered probiotic that produces 73.4 ± 47.2 nmol and 149 ± 123.6 nmol of ILA per gram of fecal and cecal matter, respectively, three days after colonization in a mouse model. In addition, hereby is reported an engineered-probiotic-related increase of ILA in the systemic circulation of the treated mice. This strain serves as proof of concept for the transfer of capacity to produce ILA in vivo and as ILA emerges as a potent microbial metabolite against gastrointestinal inflammation, further development of this strain offers efficient options for ILA-focused therapeutic interventions in situ.


Assuntos
Colite , Probióticos , Camundongos , Animais , Escherichia coli/genética , Colite/terapia , Colite/microbiologia , Fezes/microbiologia , Ceco , Indóis
3.
Nat Microbiol ; 6(11): 1367-1382, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34675385

RESUMO

Breastfeeding profoundly shapes the infant gut microbiota, which is critical for early life immune development, and the gut microbiota can impact host physiology in various ways, such as through the production of metabolites. However, few breastmilk-dependent microbial metabolites mediating host-microbiota interactions are currently known. Here, we demonstrate that breastmilk-promoted Bifidobacterium species convert aromatic amino acids (tryptophan, phenylalanine and tyrosine) into their respective aromatic lactic acids (indolelactic acid, phenyllactic acid and 4-hydroxyphenyllactic acid) via a previously unrecognized aromatic lactate dehydrogenase (ALDH). The ability of Bifidobacterium species to convert aromatic amino acids to their lactic acid derivatives was confirmed using monocolonized mice. Longitudinal profiling of the faecal microbiota composition and metabolome of Danish infants (n = 25), from birth until 6 months of age, showed that faecal concentrations of aromatic lactic acids are correlated positively with the abundance of human milk oligosaccharide-degrading Bifidobacterium species containing the ALDH, including Bifidobacterium longum, B. breve and B. bifidum. We further demonstrate that faecal concentrations of Bifidobacterium-derived indolelactic acid are associated with the capacity of these samples to activate in vitro the aryl hydrocarbon receptor (AhR), a receptor important for controlling intestinal homoeostasis and immune responses. Finally, we show that indolelactic acid modulates ex vivo immune responses of human CD4+ T cells and monocytes in a dose-dependent manner by acting as an agonist of both the AhR and hydroxycarboxylic acid receptor 3 (HCA3). Our findings reveal that breastmilk-promoted Bifidobacterium species produce aromatic lactic acids in the gut of infants and suggest that these microbial metabolites may impact immune function in early life.


Assuntos
Bifidobacterium/metabolismo , Microbioma Gastrointestinal , Ácido Láctico/metabolismo , Adulto , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Bifidobacterium/química , Bifidobacterium/classificação , Bifidobacterium/genética , Aleitamento Materno , Estudos de Coortes , Fezes/microbiologia , Feminino , Humanos , Lactente , Ácido Láctico/química , Masculino , Camundongos , Receptores de Hidrocarboneto Arílico/metabolismo , Adulto Jovem
4.
Br J Nutr ; 125(12): 1374-1385, 2021 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-32967742

RESUMO

Casein glycomacropeptide (CGMP) is a bioactive milk-derived peptide with potential anti-inflammatory effects. Animal studies suggest that CGMP may work by altering gut microbiota composition and enhancing butyrate production. Its effects on intestinal homoeostasis, microbiota and metabolites in humans are unknown. The aim of the present study was to assess both the intestinal and systemic immunomodulatory effects of orally ingested CGMP. We hypothesised that daily oral CGMP intake would reduce high-sensitive C-reactive protein (hsCRP) in healthy adults. In a single-centre limited but randomised, double-blinded, reference-controlled study, we compared the effects of a 4-week intervention of either 25 g of oral powder-based chocolate-flavoured CGMP or a reference drink. We included twenty-four healthy adults who all completed the study. CGMP had no systemic or intestinal immunomodulatory effects compared with a reference drink, with regard to either hsCRP or faecal calprotectin level, faecal microbiota composition or faecal SCFA content. CGMP ingestion did not affect satiety or body weight, and it caused no severe adverse events. The palatability of CGMP was acceptable, and adherence was high. CGMP did not induce or change gastrointestinal symptoms. In conclusion, we found no immunomodulatory effects of CGMP in healthy adults. In a minor group of healthy adults, oral ingestion of 25 g of CGMP during 4 weeks was safe, well tolerated, had acceptable palatability and was without any effects on body weight.


Assuntos
Butiratos/análise , Proteína C-Reativa/análise , Caseínas/administração & dosagem , Suplementos Nutricionais , Fezes/química , Microbioma Gastrointestinal , Fragmentos de Peptídeos/administração & dosagem , Adolescente , Adulto , Peso Corporal , Citocinas/sangue , Método Duplo-Cego , Ácidos Graxos Voláteis/análise , Fezes/microbiologia , Humanos , Pessoa de Meia-Idade , Saciação , Adulto Jovem
5.
PLoS One ; 15(9): e0238648, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32947608

RESUMO

Elevated postprandial plasma glucose is a risk factor for development of type 2 diabetes and cardiovascular disease. We hypothesized that the inter-individual postprandial plasma glucose response varies partly depending on the intestinal microbiome composition and function. We analyzed data from Danish adults (n = 106), who were self-reported healthy and attended the baseline visit of two previously reported randomized controlled cross-over trials within the Gut, Grain and Greens project. Plasma glucose concentrations at five time points were measured before and during three hours after a standardized breakfast. Based on these data, we devised machine learning algorithms integrating bio-clinical, as well as shotgun-sequencing-derived taxa and functional potentials of the intestinal microbiome to predict individual postprandial glucose excursions. In this post hoc study, we found microbial and clinical features, which predicted up to 48% of the inter-individual variance of postprandial plasma glucose responses (Pearson correlation coefficient of measured vs. predicted values, R = 0.69, 95% CI: 0.45 to 0.84, p<0.001). The features were age, fasting serum triglycerides, systolic blood pressure, BMI, fasting total serum cholesterol, abundance of Bifidobacterium genus, richness of metagenomics species and abundance of a metagenomic species annotated to Clostridiales at order level. A model based only on microbial features predicted up to 14% of the variance in postprandial plasma glucose excursions (R = 0.37, 95% CI: 0.02 to 0.64, p = 0.04). Adding fasting glycaemic measures to the model including microbial and bio-clinical features increased the predictive power to R = 0.78 (95% CI: 0.59 to 0.89, p<0.001), explaining more than 60% of the inter-individual variance of postprandial plasma glucose concentrations. The outcome of the study points to a potential role of the taxa and functional potentials of the intestinal microbiome. If validated in larger studies our findings may be included in future algorithms attempting to develop personalized nutrition, especially for prediction of individual blood glucose excursions in dys-glycaemic individuals.


Assuntos
Glicemia/metabolismo , Microbioma Gastrointestinal , Período Pós-Prandial , Algoritmos , Jejum/sangue , Feminino , Humanos , Estilo de Vida , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Fenômica
6.
Sci Rep ; 10(1): 7805, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32385373

RESUMO

Transplantation of germ-free (GF) mice with microbiota from mice or humans stimulates the intestinal immune system in disparate ways. We transplanted a human microbiota into GF C57BL/6 mice and a murine C57BL/6 microbiota into GF C57BL/6 mice and Swiss-Webster (SW) mice. Mice were bred to produce an offspring generation. 56% of the Operational Taxonomic Units (OTUs) present in the human donor microbiota established in the recipient mice, whereas 81% of the C57BL/6 OTUs established in the recipient C57BL/6 and SW mice. Anti-inflammatory bacteria such as Faecalibacterium and Bifidobacterium from humans were not transferred to mice. Expression of immune-related intestinal genes was lower in human microbiota-mice and not different between parent and offspring generation. Expression of intestinal barrier-related genes was slightly higher in human microbiota-mice. Cytokines and chemokines measured in plasma were differentially present in human and mouse microbiota-mice. Minor differences in microbiota and gene expression were found between transplanted mice of different genetics. It is concluded that important immune-regulating bacteria are lost when transplanting microbiota from humans to C57BL/6 mice, and that the established human microbiota is a weak stimulator of the murine immune system. The results are important for study design considerations in microbiota transplantation studies involving immunological parameters.


Assuntos
Bactérias/imunologia , Microbioma Gastrointestinal/imunologia , Sistema Imunitário/microbiologia , Transplantes/microbiologia , Animais , Bifidobacterium , Colo/microbiologia , Microbioma Gastrointestinal/genética , Vida Livre de Germes/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL
7.
Nat Commun ; 9(1): 3294, 2018 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-30120222

RESUMO

Accumulating evidence implicates metabolites produced by gut microbes as crucial mediators of diet-induced host-microbial cross-talk. Here, we review emerging data suggesting that microbial tryptophan catabolites resulting from proteolysis are influencing host health. These metabolites are suggested to activate the immune system through binding to the aryl hydrocarbon receptor (AHR), enhance the intestinal epithelial barrier, stimulate gastrointestinal motility, as well as secretion of gut hormones, exert anti-inflammatory, anti-oxidative or toxic effects in systemic circulation, and putatively modulate gut microbial composition. Tryptophan catabolites thus affect various physiological processes and may contribute to intestinal and systemic homeostasis in health and disease.


Assuntos
Bactérias/metabolismo , Doença , Saúde , Triptofano/metabolismo , Animais , Sistema Digestório/metabolismo , Humanos , Receptores de Hidrocarboneto Arílico/metabolismo
8.
Diabetologia ; 61(4): 810-820, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29379988

RESUMO

AIMS/HYPOTHESIS: Individuals with type 2 diabetes have aberrant intestinal microbiota. However, recent studies suggest that metformin alters the composition and functional potential of gut microbiota, thereby interfering with the diabetes-related microbial signatures. We tested whether specific gut microbiota profiles are associated with prediabetes (defined as fasting plasma glucose of 6.1-7.0 mmol/l or HbA1c of 42-48 mmol/mol [6.0-6.5%]) and a range of clinical biomarkers of poor metabolic health. METHODS: In the present case-control study, we analysed the gut microbiota of 134 Danish adults with prediabetes, overweight, insulin resistance, dyslipidaemia and low-grade inflammation and 134 age- and sex-matched individuals with normal glucose regulation. RESULTS: We found that five bacterial genera and 36 operational taxonomic units (OTUs) were differentially abundant between individuals with prediabetes and those with normal glucose regulation. At the genus level, the abundance of Clostridium was decreased (mean log2 fold change -0.64 (SEM 0.23), p adj = 0.0497), whereas the abundances of Dorea, [Ruminococcus], Sutterella and Streptococcus were increased (mean log2 fold change 0.51 (SEM 0.12), p adj = 5 × 10-4; 0.51 (SEM 0.11), p adj = 1 × 10-4; 0.60 (SEM 0.21), p adj = 0.0497; and 0.92 (SEM 0.21), p adj = 4 × 10-4, respectively). The two OTUs that differed the most were a member of the order Clostridiales (OTU 146564) and Akkermansia muciniphila, which both displayed lower abundance among individuals with prediabetes (mean log2 fold change -1.74 (SEM 0.41), p adj = 2 × 10-3 and -1.65 (SEM 0.34), p adj = 4 × 10-4, respectively). Faecal transfer from donors with prediabetes or screen-detected, drug-naive type 2 diabetes to germfree Swiss Webster or conventional C57BL/6 J mice did not induce impaired glucose regulation in recipient mice. CONCLUSIONS/INTERPRETATION: Collectively, our data show that individuals with prediabetes have aberrant intestinal microbiota characterised by a decreased abundance of the genus Clostridium and the mucin-degrading bacterium A. muciniphila. Our findings are comparable to observations in overt chronic diseases characterised by low-grade inflammation.


Assuntos
Diabetes Mellitus Tipo 2/microbiologia , Microbioma Gastrointestinal , Estado Pré-Diabético/microbiologia , Idoso , Animais , Antropometria , Biomarcadores/metabolismo , Glicemia/análise , Estudos de Casos e Controles , Dinamarca , Dislipidemias/epidemiologia , Dislipidemias/microbiologia , Feminino , Humanos , Inflamação , Resistência à Insulina , Masculino , Metformina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Estado Pré-Diabético/complicações , RNA Ribossômico 16S/metabolismo
9.
Front Microbiol ; 8: 356, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28321211

RESUMO

The establishment of the human gut microbiota in early life has been associated with later health and disease. During the 1st months after birth, the microbial composition in the gut is known to be affected by the mode of delivery, use of antibiotics, geographical location and type of feeding (breast/formula). Consequently, the neonatal period and early infancy has attracted much attention. However, after this first period the gut microbial composition continues to develop until the age of 3 years, and these 1st years have been designated "a window of opportunity" for microbial modulation. The beginning and end of this window is currently debated, but it likely coincides with the complementary feeding period, marking the gradual transition from milk-based infant feeding to family diet usually occurring between 6 and 24 months. Furthermore, the 'first 1000 days,' i.e., the period from conception until age 2 years, are generally recognized to be of particular importance for the healthy development of children. While dietary changes are known to affect the adult gut microbiota, there is a gap in our knowledge on how the introduction of new dietary components into the diet of infants/young children affects the gut microbiota development. This perspective paper summarizes the currently very few studies addressing the effects of complementary diet on gut microbiota, and highlights the recent finding that transition to family foods greatly impacts the development of gut microbial diversity. Further, we discuss potential impacts on child health and the need for further studies on this important topic.

10.
Sci Rep ; 7: 42245, 2017 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-28169374

RESUMO

Germ-free rodents colonized with microbiotas of interest are used for host-microbiota investigations and for testing microbiota-targeted therapeutic candidates. Traditionally, isolators are used for housing such gnotobiotic rodents due to optimal protection from the environment, but research groups focused on the microbiome are increasingly combining or substituting isolator housing with individually ventilated cage (IVC) systems. We compared the effect of housing systems on the gut microbiota composition of germ-free mice colonized with a complex microbiota and housed in either multiple IVC cages in an IVC facility or in multiple open-top cages in an isolator during three generations and five months. No increase in bacterial diversity as assessed by 16S rRNA gene sequencing was observed in the IVC cages, despite not applying completely aseptic cage changes. The donor bacterial community was equally represented in both housing systems. Time-dependent clustering between generations was observed in both systems, but was strongest in the IVC cages. Different relative abundance of a Rikenellaceae genus contributed to separate clustering of the isolator and IVC communities. Our data suggest that complex microbiotas are protected in IVC systems, but challenges related to temporal dynamics should be addressed.


Assuntos
Microbioma Gastrointestinal , Vida Livre de Germes , Abrigo para Animais , Ventilação , Envelhecimento/fisiologia , Animais , Biodiversidade , Análise por Conglomerados , Contagem de Colônia Microbiana , Fezes/microbiologia , Feminino , Masculino , Camundongos Endogâmicos C57BL , Filogenia , Fatores de Tempo
11.
Nat Microbiol ; 1(9): 16093, 2016 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-27562254

RESUMO

Little is known about how colonic transit time relates to human colonic metabolism and its importance for host health, although a firm stool consistency, a proxy for a long colonic transit time, has recently been positively associated with gut microbial richness. Here, we show that colonic transit time in humans, assessed using radio-opaque markers, is associated with overall gut microbial composition, diversity and metabolism. We find that a long colonic transit time associates with high microbial richness and is accompanied by a shift in colonic metabolism from carbohydrate fermentation to protein catabolism as reflected by higher urinary levels of potentially deleterious protein-derived metabolites. Additionally, shorter colonic transit time correlates with metabolites possibly reflecting increased renewal of the colonic mucosa. Together, this suggests that a high gut microbial richness does not per se imply a healthy gut microbial ecosystem and points at colonic transit time as a highly important factor to consider in microbiome and metabolomics studies.


Assuntos
Microbioma Gastrointestinal , Trânsito Gastrointestinal , Metaboloma , Adulto , Idoso , Biomarcadores/metabolismo , Metabolismo dos Carboidratos , Colo/metabolismo , Fezes/microbiologia , Feminino , Fermentação , Humanos , Masculino , Metabolismo , Pessoa de Meia-Idade , Mucosa/metabolismo , Proteínas/metabolismo , Reprodutibilidade dos Testes , Adulto Jovem
12.
PLoS One ; 10(11): e0142352, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26562532

RESUMO

OBJECTIVE: The gut microbiota has been designated as an active regulator of glucose metabolism and metabolic phenotype in a number of animal and human observational studies. We evaluated the effect of removing as many bacteria as possible by antibiotics on postprandial physiology in healthy humans. METHODS: Meal tests with measurements of postprandial glucose tolerance and postprandial release of insulin and gut hormones were performed before, immediately after and 6 weeks after a 4-day, broad-spectrum, per oral antibiotic cocktail (vancomycin 500 mg, gentamycin 40 mg and meropenem 500 mg once-daily) in a group of 12 lean and glucose tolerant males. Faecal samples were collected for culture-based assessment of changes in gut microbiota composition. RESULTS: Acute and dramatic reductions in the abundance of a representative set of gut bacteria was seen immediately following the antibiotic course, but no changes in postprandial glucose tolerance, insulin secretion or plasma lipid concentrations were found. Apart from an acute and reversible increase in peptide YY secretion, no changes were observed in postprandial gut hormone release. CONCLUSION: As evaluated by selective cultivation of gut bacteria, a broad-spectrum 4-day antibiotics course with vancomycin, gentamycin and meropenem induced shifts in gut microbiota composition that had no clinically relevant short or long-term effects on metabolic variables in healthy glucose-tolerant males. TRIAL REGISTRATION: clinicaltrials.gov NCT01633762.


Assuntos
Antibacterianos/farmacologia , Hormônios Gastrointestinais/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Glucose/metabolismo , Adolescente , Adulto , Antibacterianos/efeitos adversos , Antibacterianos/metabolismo , Carga Bacteriana , Glicemia/metabolismo , Diarreia/induzido quimicamente , Jejum/sangue , Fezes/química , Fezes/microbiologia , Hormônios Gastrointestinais/sangue , Gentamicinas/efeitos adversos , Gentamicinas/metabolismo , Gentamicinas/farmacologia , Humanos , Insulina/sangue , Masculino , Meropeném , Período Pós-Prandial , Tienamicinas/efeitos adversos , Tienamicinas/metabolismo , Tienamicinas/farmacologia , Fatores de Tempo , Resultado do Tratamento , Vancomicina/efeitos adversos , Vancomicina/metabolismo , Vancomicina/farmacologia , Adulto Jovem
13.
Immun Inflamm Dis ; 3(3): 309-20, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26417445

RESUMO

To assess the microbial influence on postnatal hematopoiesis, we examined the role of early life microbial colonization on the composition of leukocyte subsets in the neonatal spleen. A high number of CD11b(+)Gr-1(+) splenocytes present perinatally was sustained for a longer period in conventionally colonized (CONV) mice than in mono-colonized (MC) and germfree (GF) mice, and the CD4(+) T cell population established faster in CONV mice. At the day of birth, compared to GF mice, the expression of Cxcl2 was up-regulated and Arg1 down-regulated in livers of CONV mice. This coincided with lower abundance of polylobed cells in the liver of CONV mice. An earlier peak in the expression of the genes Tjp1, Cdh1, and JamA in intestinal epithelial cells of CONV mice indicated an accelerated closure of the epithelial barrier. In conclusion, we have identified an important microbiota-dependent neonatal hematopoietic event, which we suggest impacts the subsequent development of the T cell population in the murine spleen.

14.
Br J Nutr ; 114(1): 63-74, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-25992463

RESUMO

Some lipid hydrolysis products such as medium-chained NEFA (MC-NEFA), sphingosine and monoacylglycerols (MAG) possess antibacterial activity, while others, including oleic acid, are essential for the optimal growth of Lactobacillus species. Thus, changes in the concentrations of NEFA and MAG in the distal ileum and colon can potentially selectively modulate the composition of the gut microbiota, especially in early life when lipid absorption efficacy is reduced. As medium-chained fatty acids are enriched in mothers' milk, such effects may be highly relevant during gut colonisation. In the present study, we examined the effect of selected NEFA, MAG and sphingosine on the composition of faecal microbial communities derived from infants aged 2-5 months during a 24 h anaerobic in vitro fermentation. We tested lipid mixtures in the concentration range of 0-200 µm, either based on MC-NEFA (10 : 0 to 14 : 0 and MAG 12 : 0) or long-chained NEFA (LC-NEFA; 16 : 0 to 18 : 1 and MAG 16 : 0) with and without sphingosine, representing lipid hydrolysis products characteristic for intestinal hydrolysis of breast milk lipids. Ion Torrent sequencing of the bacterial 16S ribosomal RNA gene revealed that the relative abundance of lactic acid-producing genera, including Lactobacillus and Bifidobacterium, was generally increased in the presence of 50 µm or higher concentrations of MC-NEFA. For Bifidobacterium, the same effect was also observed in the presence of a mixture containing LC-NEFA with sphingosine. On the contrary, the relative abundance of Enterobacteriaceae was significantly decreased in the presence of both lipid mixtures. Our findings suggest that the high concentration of medium-chained fatty acids in breast milk might have functional effects on the establishment of the gut microbiota in early life.


Assuntos
Ácidos Graxos não Esterificados/farmacologia , Fezes/microbiologia , Mucosa Intestinal/metabolismo , Microbiota , Monoglicerídeos/farmacologia , Esfingosina/farmacologia , Bactérias/classificação , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/genética , Bifidobacterium/crescimento & desenvolvimento , DNA Bacteriano/análise , DNA Bacteriano/isolamento & purificação , Enterobacteriaceae/efeitos dos fármacos , Enterobacteriaceae/genética , Enterobacteriaceae/crescimento & desenvolvimento , Fermentação , Humanos , Hidrólise , Lactente , Lactobacillus/efeitos dos fármacos , Lactobacillus/genética , Lactobacillus/crescimento & desenvolvimento , Metabolismo dos Lipídeos , Leite Humano/química , RNA Ribossômico 16S/genética
15.
Environ Microbiol ; 17(4): 1134-51, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24920558

RESUMO

Listeria monocytogenes can cause the potentially fatal food-borne disease listeriosis, and the use of bacteriocin-producing lactic acid bacteria to control L. monocytogenes holds great promise. However, the development of bacteriocin resistance is a potential challenge, and the purpose of this study was to determine if exposure to sublethal concentrations of pediocin-containing Lactobacillus plantarum WHE 92 supernatant could prime L. monocytogenes for resistance. By transcriptomic analysis, we found two, 55 and 539 genes differentially expressed after 10, 60 and 180 min of exposure to L. plantarum WHE 92 supernatant as compared with control exposures. We observed temporal expression changes in genes regulated by the two component system LisRK and the alternative sigma factors SigB and SigL. Additionally, several genes involved in bacteriocin resistance were induced. ΔlisR, ΔsigB and ΔsigL mutants were all more resistant than wild types to L. plantarum WHE 92 supernatant. Conclusively, LisRK, SigB and SigL regulation and genes associated with resistance are involved in the temporal adaptive response to pediocin, and all three regulatory systems affect pediocin resistance. Thus, a single exposure to a sublethal pediocin concentration initiates a response pointing to resistance, and indicates that further research exploring the link between adaptive responses and resistance is needed.


Assuntos
Bacteriocinas/farmacologia , Membrana Celular/metabolismo , Farmacorresistência Bacteriana/genética , Doenças Transmitidas por Alimentos/microbiologia , Lactobacillus plantarum/metabolismo , Listeria monocytogenes/efeitos dos fármacos , Listeriose/microbiologia , Fator sigma/genética , Adaptação Fisiológica/genética , Parede Celular/efeitos dos fármacos , Parede Celular/genética , Humanos , Lactobacillus plantarum/genética , Listeria monocytogenes/genética , Pediocinas , Fator sigma/metabolismo , Estresse Fisiológico
16.
Sci Rep ; 4: 5922, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25082483

RESUMO

Transferring gut microbiota from one individual to another may enable researchers to "humanize" the gut of animal models and transfer phenotypes between species. To date, most studies of gut microbiota transfer are performed in germ-free mice. In the studies presented, it was tested whether an antibiotic treatment approach could be used instead. C57BL/6 mice were treated with ampicillin prior to inoculation at weaning or eight weeks of age with gut microbiota from lean or obese donors. The gut microbiota and clinical parameters of the recipients was characterized one and six weeks after inoculation. The results demonstrate, that the donor gut microbiota was introduced, established, and changed the gut microbiota of the recipients. Six weeks after inoculation, the differences persisted, however alteration of the gut microbiota occurred with time within the groups. The clinical parameters of the donor phenotype were partly transmissible from obese to lean mice, in particularly ß cell hyperactivity in the obese recipients. Thus, a successful inoculation of gut microbiota was not age dependent in order for the microbes to colonize, and transferring different microbial compositions to conventional antibiotic-treated mice was possible at least for a time period during which the microbiota may permanently modulate important host functions.


Assuntos
Ampicilina/farmacologia , Antibacterianos/farmacologia , Trato Gastrointestinal/microbiologia , Microbiota , Animais , Feminino , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Obesos
17.
Gut Microbes ; 5(3): 296-303, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24717228

RESUMO

Monocolonization of germ-free (GF) mice enables the study of specific bacterial species in vivo. Lactobacillus acidophilus NCFM(TM) (NCFM) is a probiotic strain; however, many of the mechanisms behind its health-promoting effect remain unknown. Here, we studied the effects of NCFM on the metabolome of jejunum, cecum, and colon of NCFM monocolonized (MC) and GF mice using liquid chromatography coupled to mass-spectrometry (LC-MS). The study adds to existing evidence that NCFM in vivo affects the bile acid signature of mice, in particular by deconjugation. Furthermore, we confirmed that carbohydrate metabolism is affected by NCFM in the mouse intestine as especially the digestion of oligosaccharides (penta- and tetrasaccharides) was increased in MC mice. Additionally, levels of α-tocopherol acetate (vitamin E acetate) were higher in the intestine of GF mice than in MC mice, suggesting that NCFM affects the vitamin E acetate metabolism. NCFM did not digest vitamin E acetate in vitro, suggesting that direct bacterial metabolism was not the cause of the altered metabolome in vivo. Taken together, our results suggest that NCFM affects intestinal carbohydrate metabolism, bile acid metabolism and vitamin E metabolism, although it remains to be investigated whether this effect is unique to NCFM.


Assuntos
Acetatos/metabolismo , Ácidos e Sais Biliares/análise , Bile/química , Intestinos/microbiologia , Lactobacillus acidophilus/crescimento & desenvolvimento , Lactobacillus acidophilus/metabolismo , Vitamina E/metabolismo , Animais , Cromatografia Líquida , Vida Livre de Germes , Intestinos/química , Espectrometria de Massas , Metaboloma , Camundongos , Probióticos/administração & dosagem
18.
Appl Environ Microbiol ; 80(3): 1142-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24296500

RESUMO

It has been suggested that the human gut microbiota can be divided into enterotypes based on the abundance of specific bacterial groups; however, the biological significance and stability of these enterotypes remain unresolved. Here, we demonstrated that subjects (n = 62) 18 to 65 years old with central obesity and components of metabolic syndrome could be grouped into two discrete groups simply by their relative abundance of Prevotella spp. divided by Bacteroides spp. (P/B ratio) obtained by quantitative PCR analysis. Furthermore, we showed that these groups remained stable during a 6-month, controlled dietary intervention, where the effect of consuming a diet in accord with the new Nordic diet (NND) recommendations as opposed to consuming the average Danish diet (ADD) on the gut microbiota was investigated. In this study, subjects (with and without stratification according to P/B ratio) did not reveal significant changes in 35 selected bacterial taxa quantified by quantitative PCR (ADD compared to NND) resulting from the dietary interventions. However, we found higher total plasma cholesterol within the high-P/B group than in the low-P/B group after the intervention. We propose that stratification of humans based simply on their P/B ratio could allow better assessment of possible effects of interventions on the gut microbiota and physiological biomarkers.


Assuntos
Bacteroides/isolamento & purificação , Dieta/métodos , Trato Gastrointestinal/microbiologia , Prevotella/isolamento & purificação , Colesterol/sangue , Fezes/microbiologia , Humanos , Metagenoma , Plasma/química , Reação em Cadeia da Polimerase em Tempo Real
19.
Nutrients ; 5(9): 3740-56, 2013 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-24064573

RESUMO

High dietary intakes of some protein sources, including soy protein, can increase colonic DNA damage in animals, whereas some carbohydrates attenuate this. We investigated whether inulin and xylo-oligosaccharides (XOS) could be protective against DNA strand breaks by adding them to a human colonic simulator consisting of a proximal vessel (PV) (pH 5.5) and a distal vessel (DV) (pH 6.8) inoculated with human faeces and media containing soy protein. Genotoxicity of the liquid phase and microbial population changes in the vessels were measured. Soy protein (3%) was fermented with 1% low amylose cornstarch for 10 day followed by soy protein with 1% XOS or 1% inulin for 10 day. Inulin did not alter genotoxicity but XOS significantly reduced PV genotoxicity and increased DV genotoxicity. Inulin and XOS significantly increased butyrate concentration in the DV but not PV. Numbers of the key butyrate-producing bacterium Faecalibacterium prausnitzii were significantly increased in the PV and DV by inulin but significantly decreased by XOS in both vessels. Other bacteria examined were also significantly impacted by the carbohydrate treatments or by the vessel (i.e., pH). There was a significant overall inverse correlation between levels of damage induced by the ferments and levels of sulphate-reducing bacteria, Bacteroides fragilis, and acetate. In conclusion, dietary XOS can potentially modulate the genotoxicity of the colonic environment and specific bacterial groups and short chain fatty acids may mediate this.


Assuntos
Bactérias/metabolismo , Colo/microbiologia , Dano ao DNA/efeitos dos fármacos , Inulina/farmacologia , Oligossacarídeos/farmacologia , Proteínas de Soja/metabolismo , Bifidobacterium/metabolismo , Butiratos/metabolismo , Colo/metabolismo , Ensaio Cometa , Ácidos Graxos Voláteis/metabolismo , Fezes/microbiologia , Fermentação , Células HT29 , Humanos
20.
Food Funct ; 4(5): 784-93, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23580006

RESUMO

The present study aimed at examining oligosaccharides (OS) for potential stimulation of probiotic bacteria. Nineteen structurally well-defined candidate OS covering groups of ß-glucosides, α-glucosides and α-galactosides with degree of polymerization 2-4 were prepared in >100 mg amounts by chemoenzymatic synthesis (i.e. reverse phosphorolysis or transglycosylation). Fourteen of the OS are not naturally occurring and five (ß-D-glucosyl-fructose, ß-D-glucosyl-xylitol, α-glucosyl-(1,4)-D-mannose, α-glucosyl-(1,4)-D-xylose; α-glucosyl-(1,4)-L-fucose) have recently been synthesized for the first time. These OS have not been previously tested for effects of bacterial growth and here the ability of all 19 OS to support growth of four gastrointestinal bacteria: three probiotic bacteria Bifidobacterium lactis, Bifidobacterium longum, and Lactobacillus acidophilus, and one commensal bacterium, Bacteroides vulgatus has been evaluated in monocultures. The disaccharides ß-D-glucosyl-xylitol and ß-D-glucosyl-(1,4)-xylose noticeably stimulated growth yields of L. acidophilus NCFM, and additionally, ß-D-glucosyl-(1,4)-xylose stimulated B. longum Bl-05. α-Glucosyl-(1,4)-glucosamine and α-glucosyl-(1,4)-N-acetyl-glucosamine enhanced the growth rate of B. animalis subsp. lactis and B. longum Bl-05, whereas L. acidophilus NCFM and Bac. vulgatus did not grow on these OS. α-Galactosyl-(1,6)-α-galactosyl-(1,6)-glucose advanced the growth rate of B. animalis subsp. lactis and L. acidophilus NCFM. Thus several of the structurally well-defined OS supported growth of beneficial gut bacteria. This reflects a broad specificity of their sugar transporters for OS, including specificity for non-naturally occurring OS, hence showing promise for design of novel prebiotics.


Assuntos
Bacteroides/crescimento & desenvolvimento , Bifidobacterium/crescimento & desenvolvimento , Trato Gastrointestinal/microbiologia , Lactobacillus acidophilus/crescimento & desenvolvimento , Oligossacarídeos/química , Bacteroides/isolamento & purificação , Bifidobacterium/isolamento & purificação , Dissacarídeos/metabolismo , Humanos , Lactobacillus acidophilus/isolamento & purificação , Manose/metabolismo , Prebióticos/análise , Probióticos , Xilose/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...