Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
EClinicalMedicine ; 61: 102070, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37528843

RESUMO

Background: Vaginal dysbiosis covers imbalances in the vaginal microbiota, defined by altered composition of bacteria, viruses, and fungi and is associated with euploid pregnancy losses, premature birth, infertility, or bacterial vaginosis. A large proportion of women who have vaginal dysbiosis do not experience any symptoms. Antibiotics are the traditional treatment, recently combined with local probiotics in some cases. Vaginal Microbiota Transplantation (VMT) with eubiotic vaginal bacterial microbiota after antibiotic eradication of pathogens has successfully been performed in a case study with five patients, but no VMT has been performed without the use of antibiotics. Methods: This is a proof of concept case study. The patient was found to have vaginal dysbiosis at the RPL clinic at Copenhagen University Hospital Hvidovre, Denmark on the 23rd of June 2021. She was offered and accepted to receive experimental treatment in the form of a VMT as a compassionate use case. VMT is the transfer of cervicovaginal secretions (CVS) from a healthy donor with a Lactobacillus-dominant vaginal microbiome to a recipient with a dysbiotic vaginal microbiome. CVS is a mixture of e.g., mucus, bacteria, metabolites present in the vaginal canal. Potential donors were thoroughly screened for the absence of STIs, and the most suitable donor sample for the specific patient in this study was determined via an in vitro microbiome competition assay. Findings: A 30-year-old patient with one livebirth and a complicated pregnancy history of two stillbirths and 1 s trimester pregnancy loss in gestational weeks 27 (2019), 17 (2020) and 23 (2020) respectively with complaints of vaginal irritation and discharge that had aggravated in all her pregnancies. Her vaginal microbiome composition showed a 90% dominance of Gardnerella spp. After one VMT there was a complete shift in microbiome composition to 81.2% L. crispatus and 9% L. jensenii with a concurrent resolvement of vaginal symptoms. Single nucleotide polymorphism-analysis confirmed her microbiome to be of donor origin and it remain stable now 1.5 years after the VMT. Five months after the VMT she became pregnant and has successfully delivered a healthy baby at term. Interpretation: Here we report a successful VMT with confirmed donor strain engraftment followed by a successful pregnancy and delivery after a series of late pregnancy losses/stillbirths. Findings suggest that VMT is a potential treatment for severe vaginal dysbiosis. Further, larger studies are required. Funding: The study was partially funded (i.e., analysis costs) by Freya Biosciences Aps, Fruebjergvej, 2100 Copenhagen, Denmark.

2.
Nat Commun ; 13(1): 1244, 2022 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-35273143

RESUMO

Relative abundances of bacterial species in the gut microbiome have been linked to many diseases. Species of gut bacteria are ecologically differentiated by their abilities to metabolize different glycans, making glycan delivery a powerful way to alter the microbiome to promote health. Here, we study the properties and therapeutic potential of chemically diverse synthetic glycans (SGs). Fermentation of SGs by gut microbiome cultures results in compound-specific shifts in taxonomic and metabolite profiles not observed with reference glycans, including prebiotics. Model enteric pathogens grow poorly on most SGs, potentially increasing their safety for at-risk populations. SGs increase survival, reduce weight loss, and improve clinical scores in mouse models of colitis. Synthetic glycans are thus a promising modality to improve health through selective changes to the gut microbiome.


Assuntos
Colite , Microbioma Gastrointestinal , Animais , Bactérias/metabolismo , Colite/tratamento farmacológico , Promoção da Saúde , Camundongos , Polissacarídeos/metabolismo
3.
Gastroenterology ; 157(3): 637-646.e4, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31095949

RESUMO

BACKGROUND & AIMS: Enteropathy and small-intestinal ulcers are common adverse effects of nonsteroidal anti-inflammatory drugs such as acetylsalicylic acid (ASA). Safe, cytoprotective strategies are needed to reduce this risk. Specific bifidobacteria might have cytoprotective activities, but little is known about these effects in humans. We used serial video capsule endoscopy (VCE) to assess the efficacy of a specific Bifidobacterium strain in healthy volunteers exposed to ASA. METHODS: We performed a single-site, double-blind, parallel-group, proof-of-concept analysis of 75 heathy volunteers given ASA (300 mg) daily for 6 weeks, from July 31 through October 24, 2017. The participants were randomly assigned (1:1) to groups given oral capsules of Bifidobacterium breve (Bif195) (≥5 × 1010 colony-forming units) or placebo daily for 8 weeks. Small-intestinal damage was analyzed by serial VCE at 6 visits. The area under the curve (AUC) for intestinal damage (Lewis score) and the AUC value for ulcers were the primary and first-ranked secondary end points of the trial, respectively. RESULTS: Efficacy data were obtained from 35 participants given Bif195 and 31 given placebo. The AUC for Lewis score was significantly lower in the Bif195 group (3040 ± 1340 arbitrary units) than the placebo group (4351 ± 3195) (P = .0376). The AUC for ulcer number was significantly lower in the Bif195 group (50.4 ± 53.1 arbitrary units) than in the placebo group (75.2 ± 85.3 arbitrary units) (P = .0258). Twelve adverse events were reported from the Bif195 group and 20 from the placebo group. None of the events was determined to be related to Bif195 intake. CONCLUSIONS: In a randomized, double-blind trial of healthy volunteers, we found oral Bif195 to safely reduce the risk of small-intestinal enteropathy caused by ASA. ClinicalTrials.gov no: NCT03228589.


Assuntos
Anti-Inflamatórios não Esteroides/efeitos adversos , Aspirina/efeitos adversos , Bifidobacterium breve/crescimento & desenvolvimento , Microbioma Gastrointestinal , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/microbiologia , Probióticos/administração & dosagem , Úlcera/prevenção & controle , Adolescente , Adulto , Endoscopia por Cápsula , Método Duplo-Cego , Feminino , Voluntários Saudáveis , Humanos , Intestino Delgado/patologia , Irlanda , Masculino , Probióticos/efeitos adversos , Fatores de Tempo , Úlcera/induzido quimicamente , Úlcera/microbiologia , Úlcera/patologia , Adulto Jovem
4.
Curr Opin Biotechnol ; 56: 55-60, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30296737

RESUMO

The different levels of knowledge described in a translational pipeline (the connection of molecular mechanisms with pre-clinical physiological and human health effects) are not complete for many probiotics. At present, we are not in a position to fully understand the mechanistic basis of many well established probiotic health benefits which, in turn, limits our ability to use mechanisms to predict which probiotics are likely to be effective in any given population. Here we suggest that this concept of a translation pipeline connecting mechanistic insights to probiotic efficacy can support the selection and production of improved probiotic products. Such a conceptual pipeline would also provide a framework for the design of clinical trials to convincingly demonstrate the benefit of probiotics to human health in well-defined subpopulations.


Assuntos
Probióticos/metabolismo , Pesquisa Translacional Biomédica , Animais , Microbioma Gastrointestinal , Humanos , Lactose/metabolismo , Probióticos/administração & dosagem , Iogurte/microbiologia
6.
Nat Commun ; 9(1): 2802, 2018 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-30022049

RESUMO

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


Assuntos
Bilophila/patogenicidade , Infecções por Desulfovibrionaceae/microbiologia , Gorduras na Dieta/efeitos adversos , Fígado Gorduroso/microbiologia , Lacticaseibacillus rhamnosus/fisiologia , Síndrome Metabólica/microbiologia , Probióticos/farmacologia , Animais , Bilophila/crescimento & desenvolvimento , Glicemia/metabolismo , Citocinas/biossíntese , Citocinas/genética , Infecções por Desulfovibrionaceae/etiologia , Infecções por Desulfovibrionaceae/metabolismo , Infecções por Desulfovibrionaceae/terapia , Dieta Hiperlipídica/efeitos adversos , Fígado Gorduroso/etiologia , Fígado Gorduroso/metabolismo , Fígado Gorduroso/terapia , Microbioma Gastrointestinal , Fígado/microbiologia , Fígado/patologia , Testes de Função Hepática , Masculino , Redes e Vias Metabólicas/genética , Síndrome Metabólica/etiologia , Síndrome Metabólica/metabolismo , Síndrome Metabólica/terapia , Camundongos , Camundongos Endogâmicos C57BL , Transcriptoma
7.
Sci Rep ; 8(1): 5098, 2018 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-29572473

RESUMO

Enterococci, in particular vancomycin-resistant enterococci (VRE), are a leading cause of hospital-acquired infections. Promoting intestinal resistance against enterococci could reduce the risk of VRE infections. We investigated the effects of two Lactobacillus strains to prevent intestinal VRE. We used an intestinal colonisation mouse model based on an antibiotic-induced microbiota dysbiosis to mimic enterococci overgrowth and VRE persistence. Each Lactobacillus spp. was administered daily to mice starting one week before antibiotic treatment until two weeks after antibiotic and VRE inoculation. Of the two strains, Lactobacillus paracasei CNCM I-3689 decreased significantly VRE numbers in the feces demonstrating an improvement of the reduction of VRE. Longitudinal microbiota analysis showed that supplementation with L. paracasei CNCM I-3689 was associated with a better recovery of members of the phylum Bacteroidetes. Bile salt analysis and expression analysis of selected host genes revealed increased level of lithocholate and of ileal expression of camp (human LL-37) upon L. paracasei CNCM I-3689 supplementation. Although a direct effect of L. paracasei CNCM I-3689 on the VRE reduction was not ruled out, our data provide clues to possible anti-VRE mechanisms supporting an indirect anti-VRE effect through the gut microbiota. This work sustains non-antibiotic strategies against opportunistic enterococci after antibiotic-induced dysbiosis.


Assuntos
Bacteroidetes/fisiologia , Lacticaseibacillus paracasei/fisiologia , Probióticos/administração & dosagem , Enterococos Resistentes à Vancomicina/fisiologia , Animais , Antibacterianos/farmacologia , Bacteroidetes/efeitos dos fármacos , Clindamicina/farmacologia , Fezes/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Intestinos/microbiologia , Masculino , Camundongos , Probióticos/farmacologia , Enterococos Resistentes à Vancomicina/efeitos dos fármacos
8.
Nat Biotechnol ; 35(11): 1069-1076, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28967887

RESUMO

Technical variation in metagenomic analysis must be minimized to confidently assess the contributions of microbiota to human health. Here we tested 21 representative DNA extraction protocols on the same fecal samples and quantified differences in observed microbial community composition. We compared them with differences due to library preparation and sample storage, which we contrasted with observed biological variation within the same specimen or within an individual over time. We found that DNA extraction had the largest effect on the outcome of metagenomic analysis. To rank DNA extraction protocols, we considered resulting DNA quantity and quality, and we ascertained biases in estimates of community diversity and the ratio between Gram-positive and Gram-negative bacteria. We recommend a standardized DNA extraction method for human fecal samples, for which transferability across labs was established and which was further benchmarked using a mock community of known composition. Its adoption will improve comparability of human gut microbiome studies and facilitate meta-analyses.


Assuntos
Fracionamento Químico/métodos , DNA/química , Fezes/química , Metagenômica , Bactérias/genética , Biologia Computacional , Humanos , Controle de Qualidade , Especificidade da Espécie
9.
Psychosom Med ; 79(8): 905-913, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28661940

RESUMO

OBJECTIVE: Brain-gut-microbiota interactions may play an important role in human health and behavior. Although rodent models have demonstrated effects of the gut microbiota on emotional, nociceptive, and social behaviors, there is little translational human evidence to date. In this study, we identify brain and behavioral characteristics of healthy women clustered by gut microbiota profiles. METHODS: Forty women supplied fecal samples for 16S rRNA profiling. Microbial clusters were identified using Partitioning Around Medoids. Functional magnetic resonance imaging was acquired. Microbiota-based group differences were analyzed in response to affective images. Structural and diffusion tensor imaging provided gray matter metrics (volume, cortical thickness, mean curvature, surface area) as well as fiber density between regions. A sparse Partial Least Square-Discrimination Analysis was applied to discriminate microbiota clusters using white and gray matter metrics. RESULTS: Two bacterial genus-based clusters were identified, one with greater Bacteroides abundance (n = 33) and one with greater Prevotella abundance (n = 7). The Prevotella group showed less hippocampal activity viewing negative valences images. White and gray matter imaging discriminated the two clusters, with accuracy of 66.7% and 87.2%, respectively. The Prevotella cluster was associated with differences in emotional, attentional, and sensory processing regions. For gray matter, the Bacteroides cluster showed greater prominence in the cerebellum, frontal regions, and the hippocampus. CONCLUSIONS: These results support the concept of brain-gut-microbiota interactions in healthy humans. Further examination of the interaction between gut microbes, brain, and affect in humans is needed to inform preclinical reports that microbial modulation may affect mood and behavior.


Assuntos
Afeto/fisiologia , Bacteroides , Microbioma Gastrointestinal , Substância Cinzenta/diagnóstico por imagem , Hipocampo/diagnóstico por imagem , Hipocampo/fisiologia , Prevotella , Substância Branca/diagnóstico por imagem , Adolescente , Adulto , Bacteroides/genética , Bacteroides/isolamento & purificação , Imagem de Tensor de Difusão , Fezes/microbiologia , Feminino , Humanos , Imageamento por Ressonância Magnética , Pessoa de Meia-Idade , Prevotella/genética , Prevotella/isolamento & purificação , RNA Ribossômico 16S/classificação , Adulto Jovem
10.
Front Microbiol ; 7: 608, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27199937

RESUMO

Growing evidence supports the efficacy of many probiotic strains in the management of gastrointestinal disorders associated with deregulated intestinal barrier function and/or structure. In particular, bifidobacteria have been studied for their efficacy to both prevent and treat a broad spectrum of animal and/or human gut disorders. The aim of the current work was thus to evaluate effects on intestinal barrier function of Bifidobacterium animalis ssp. lactis CNCM-I2494, a strain used in fermented dairy products. A chronic dinitrobenzene sulfonic acid (DNBS)-induced low-grade inflammation model causing gut dysfunction in mice was used in order to study markers of inflammation, intestinal permeability, and immune function in the presence of the bacterial strain. In this chronic low-grade inflammation mice model several parameters pointed out the absence of an over active inflammation process. However, gut permeability, lymphocyte populations, and colonic cytokines were found to be altered. B. animalis ssp. lactis CNCM-I2494 was able to protect barrier functions by restoring intestinal permeability, colonic goblet cell populations, and cytokine levels. Furthermore, tight junction (TJ) proteins levels were also measured by qRT-PCR showing the ability of this strain to specifically normalize the level of several TJ proteins, in particular for claudin-4. Finally, B. lactis strain counterbalanced CD4(+) lymphocyte alterations in both spleen and mesenteric lymphoid nodes. It restores the Th1/Th2 ratio altered by the DNBS challenge (which locally augments CD4(+) Th1 cells) by increasing the Th2 response as measured by the increase in the production of major representative Th2 cytokines (IL-4, IL-5, and IL-10). Altogether, these data suggest that B. animalis ssp. lactis CNCM-I2494 may efficiently prevent disorders associated with increased barrier permeability.

11.
ISME J ; 10(9): 2235-45, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26953599

RESUMO

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


Assuntos
Bactérias/metabolismo , Produtos Fermentados do Leite/microbiologia , Disbiose/terapia , Microbioma Gastrointestinal , Lactococcus lactis/fisiologia , Animais , Bactérias/genética , Disbiose/microbiologia , Ecologia , Fezes/microbiologia , Vida Livre de Germes , Masculino , Ratos , Análise de Sequência de DNA
12.
PLoS One ; 10(10): e0138880, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26439630

RESUMO

BACKGROUND AND OBJECTIVES: Identification of new targets for metabolic diseases treatment or prevention is required. In this context, FIAF/ANGPTL4 appears as a crucial regulator of energy homeostasis. Lactobacilli are often considered to display beneficial effect for their hosts, acting on different regulatory pathways. The aim of the present work was to study the effect of several lactobacilli strains on Fiaf gene expression in human intestinal epithelial cells (IECs) and on mice tissues to decipher the underlying mechanisms. SUBJECTS AND METHODS: Nineteen lactobacilli strains have been tested on HT-29 human intestinal epithelial cells for their ability to regulate Fiaf gene expression by RT-qPCR. In order to determine regulated pathways, we analysed the whole genome transcriptome of IECs. We then validated in vivo bacterial effects using C57BL/6 mono-colonized mice fed with normal chow. RESULTS: We identified one strain (Lactobacillus rhamnosus CNCMI-4317) that modulated Fiaf expression in IECs. This regulation relied potentially on bacterial surface-exposed molecules and seemed to be PPAR-γ independent but PPAR-α dependent. Transcriptome functional analysis revealed that multiple pathways including cellular function and maintenance, lymphoid tissue structure and development, as well as lipid metabolism were regulated by this strain. The regulation of immune system and lipid and carbohydrate metabolism was also confirmed by overrepresentation of Gene Ontology terms analysis. In vivo, circulating FIAF protein was increased by the strain but this phenomenon was not correlated with modulation Fiaf expression in tissues (except a trend in distal small intestine). CONCLUSION: We showed that Lactobacillus rhamnosus CNCMI-4317 induced Fiaf expression in human IECs, and increased circulating FIAF protein level in mice. Moreover, this effect was accompanied by transcriptome modulation of several pathways including immune response and metabolism in vitro.


Assuntos
Células Epiteliais/metabolismo , Intestinos/citologia , Lacticaseibacillus rhamnosus/fisiologia , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/genética , Angiopoietinas/metabolismo , Animais , Células HT29 , Humanos , Lactobacillus/fisiologia , Camundongos , Camundongos Endogâmicos C57BL
13.
Proc Natl Acad Sci U S A ; 112(25): 7803-8, 2015 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-26056274

RESUMO

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


Assuntos
Colite/metabolismo , Lactococcus lactis/metabolismo , Muramidase/metabolismo , Superóxido Dismutase/metabolismo , Animais , Colite/enzimologia , Colite/microbiologia , Mucosa Intestinal/enzimologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Camundongos , Espécies Reativas de Oxigênio/metabolismo
14.
Trends Microbiol ; 23(6): 354-66, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25840765

RESUMO

The human gut contains a highly diverse microbial community that is essentially an open ecosystem, despite being deeply embedded within the human body. Food-associated fermentative bacteria, including probiotics, are major sources of ingested bacteria that may temporarily complement resident microbial communities, thus forming part of our transient microbiome. Here, we review data on the fate and activity of ingested bacteria and, in particular, lactobacilli and bifidobacteria in the gastrointestinal (GI) tract and their impact on the composition and metabolism of the gut microbiome with a focus on data from clinical studies. In addition, we discuss the mechanisms involved and the potential impact on the host's health.


Assuntos
Ingestão de Alimentos , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Interações Microbianas , Probióticos , Fenômenos Fisiológicos Bacterianos , Bifidobacterium/fisiologia , Ensaios Clínicos como Assunto , Dieta , Ácidos Graxos Voláteis/biossíntese , Trato Gastrointestinal/anatomia & histologia , Humanos , Lactobacillus/fisiologia , Probióticos/administração & dosagem , Probióticos/metabolismo , Propionibacteriaceae/fisiologia
15.
ISME J ; 9(1): 1-15, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24936764

RESUMO

Structural disruption of gut microbiota and associated inflammation are considered important etiological factors in high fat diet (HFD)-induced metabolic syndrome (MS). Three candidate probiotic strains, Lactobacillus paracasei CNCM I-4270 (LC), L. rhamnosus I-3690 (LR) and Bifidobacterium animalis subsp. lactis I-2494 (BA), were individually administered to HFD-fed mice (10(8) cells day(-1)) for 12 weeks. Each strain attenuated weight gain and macrophage infiltration into epididymal adipose tissue and markedly improved glucose-insulin homeostasis and hepatic steatosis. Weighted UniFrac principal coordinate analysis based on 454 pyrosequencing of fecal bacterial 16S rRNA genes showed that the probiotic strains shifted the overall structure of the HFD-disrupted gut microbiota toward that of lean mice fed a normal (chow) diet. Redundancy analysis revealed that abundances of 83 operational taxonomic units (OTUs) were altered by probiotics. Forty-nine altered OTUs were significantly correlated with one or more host MS parameters and were designated 'functionally relevant phylotypes'. Thirteen of the 15 functionally relevant OTUs that were negatively correlated with MS phenotypes were promoted, and 26 of the 34 functionally relevant OTUs that were positively correlated with MS were reduced by at least one of the probiotics, but each strain changed a distinct set of functionally relevant OTUs. LC and LR increased cecal acetate but did not affect circulating lipopolysaccharide-binding protein; in contrast, BA did not increase acetate but significantly decreased adipose and hepatic tumor necrosis factor-α gene expression. These results suggest that Lactobacillus and Bifidobacterium differentially attenuate obesity comorbidities in part through strain-specific impacts on MS-associated phylotypes of gut microbiota in mice.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Trato Gastrointestinal/microbiologia , Síndrome Metabólica/terapia , Microbiota , Probióticos/uso terapêutico , Animais , Bactérias/classificação , Bactérias/isolamento & purificação , Bifidobacterium , Fezes/microbiologia , Inflamação/terapia , Intestinos/microbiologia , Lactobacillus , Masculino , Síndrome Metabólica/etiologia , Síndrome Metabólica/microbiologia , Camundongos , Camundongos Endogâmicos C57BL
16.
Sci Rep ; 4: 6328, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25209713

RESUMO

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


Assuntos
Produtos Fermentados do Leite , Síndrome do Intestino Irritável/microbiologia , Microbiota/genética , Probióticos , Estômago/microbiologia , Bifidobacterium/crescimento & desenvolvimento , Bilophila/crescimento & desenvolvimento , Butiratos/metabolismo , Dieta , Fezes/microbiologia , Microbiologia de Alimentos , Humanos , Lactobacillus delbrueckii/crescimento & desenvolvimento , Lactococcus lactis/crescimento & desenvolvimento , RNA Ribossômico 16S/genética , Streptococcus thermophilus/crescimento & desenvolvimento
17.
PLoS One ; 9(4): e94875, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24743599

RESUMO

Bifidobacterium animalis subsp. lactis CNCM I-2494 is a component of a commercialized fermented dairy product for which beneficial effects on health has been studied by clinical and preclinical trials. To date little is known about the molecular mechanisms that could explain the beneficial effects that bifidobacteria impart to the host. Restriction-modification (R-M) systems have been identified as key obstacles in the genetic accessibility of bifidobacteria, and circumventing these is a prerequisite to attaining a fundamental understanding of bifidobacterial attributes, including the genes that are responsible for health-promoting properties of this clinically and industrially important group of bacteria. The complete genome sequence of B. animalis subsp. lactis CNCM I-2494 is predicted to harbour the genetic determinants for two type II R-M systems, designated BanLI and BanLII. In order to investigate the functionality and specificity of these two putative R-M systems in B. animalis subsp. lactis CNCM I-2494, we employed PacBio SMRT sequencing with associated methylome analysis. In addition, the contribution of the identified R-M systems to the genetic accessibility of this strain was assessed.


Assuntos
Bifidobacterium/genética , Metilação de DNA , Genômica/métodos , Mapeamento por Restrição/métodos , Análise de Sequência/métodos , Sequência de Bases , Escherichia coli/genética , Genoma Bacteriano/genética , Plasmídeos/genética
18.
ISME J ; 8(7): 1403-17, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24500617

RESUMO

Dysregulated immune responses to gut microbes are central to inflammatory bowel disease (IBD), and gut microbial activity can fuel chronic inflammation. Examining how IBD-directed therapies influence gut microbiomes may identify microbial community features integral to mitigating disease and maintaining health. However, IBD patients often receive multiple treatments during disease flares, confounding such analyses. Preclinical models of IBD with well-defined disease courses and opportunities for controlled treatment exposures provide a valuable solution. Here, we surveyed the gut microbiome of the T-bet(-/-) Rag2(-/-) mouse model of colitis during active disease and treatment-induced remission. Microbial features modified among these conditions included altered potential for carbohydrate and energy metabolism and bacterial pathogenesis, specifically cell motility and signal transduction pathways. We also observed an increased capacity for xenobiotics metabolism, including benzoate degradation, a pathway linking host adrenergic stress with enhanced bacterial virulence, and found decreased levels of fecal dopamine in active colitis. When transferred to gnotobiotic mice, gut microbiomes from mice with active disease versus treatment-induced remission elicited varying degrees of colitis. Thus, our study provides insight into specific microbial clades and pathways associated with health, active disease and treatment interventions in a mouse model of colitis.


Assuntos
Colite/microbiologia , Trato Gastrointestinal/microbiologia , Doenças Inflamatórias Intestinais/microbiologia , Microbiota/genética , Animais , Antibacterianos/farmacologia , Ácido Benzoico/metabolismo , Metabolismo dos Carboidratos , Movimento Celular , Colite/tratamento farmacológico , Colite/genética , Colite/patologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Dopamina/metabolismo , Metabolismo Energético , Humanos , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/microbiologia , Inflamação/patologia , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Camundongos , Camundongos Knockout , Microbiota/efeitos dos fármacos , Filogenia , RNA Ribossômico 16S/classificação , RNA Ribossômico 16S/genética , Indução de Remissão , Transdução de Sinais , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética
19.
PLoS One ; 8(7): e68731, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23894338

RESUMO

Lactobacillus paracasei is a member of the normal human and animal gut microbiota and is used extensively in the food industry in starter cultures for dairy products or as probiotics. With the development of low-cost, high-throughput sequencing techniques it has become feasible to sequence many different strains of one species and to determine its "pan-genome". We have sequenced the genomes of 34 different L. paracasei strains, and performed a comparative genomics analysis. We analysed genome synteny and content, focussing on the pan-genome, core genome and variable genome. Each genome was shown to contain around 2800-3100 protein-coding genes, and comparative analysis identified over 4200 ortholog groups that comprise the pan-genome of this species, of which about 1800 ortholog groups make up the conserved core. Several factors previously associated with host-microbe interactions such as pili, cell-envelope proteinase, hydrolases p40 and p75 or the capacity to produce short branched-chain fatty acids (bkd operon) are part of the L. paracasei core genome present in all analysed strains. The variome consists mainly of hypothetical proteins, phages, plasmids, transposon/conjugative elements, and known functions such as sugar metabolism, cell-surface proteins, transporters, CRISPR-associated proteins, and EPS biosynthesis proteins. An enormous variety and variability of sugar utilization gene cassettes were identified, with each strain harbouring between 25-53 cassettes, reflecting the high adaptability of L. paracasei to different niches. A phylogenomic tree was constructed based on total genome contents, and together with an analysis of horizontal gene transfer events we conclude that evolution of these L. paracasei strains is complex and not always related to niche adaptation. The results of this genome content comparison was used, together with high-throughput growth experiments on various carbohydrates, to perform gene-trait matching analysis, in order to link the distribution pattern of a specific phenotype to the presence/absence of specific sets of genes.


Assuntos
Variação Genética , Genoma Bacteriano , Genômica , Lactobacillus/genética , Metabolismo dos Carboidratos/genética , Análise por Conglomerados , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Ácidos Graxos/metabolismo , Ordem dos Genes , Transferência Genética Horizontal , Lactobacillus/classificação , Lactobacillus/metabolismo , Anotação de Sequência Molecular , Filogenia , Plasmídeos/genética
20.
Gastroenterology ; 143(4): 913-6.e7, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22728514

RESUMO

Alterations in intestinal microbiota are associated with obesity and insulin resistance. We studied the effects of infusing intestinal microbiota from lean donors to male recipients with metabolic syndrome on the recipients' microbiota composition and glucose metabolism. Subjects were assigned randomly to groups that were given small intestinal infusions of allogenic or autologous microbiota. Six weeks after infusion of microbiota from lean donors, insulin sensitivity of recipients increased (median rate of glucose disappearance changed from 26.2 to 45.3 µmol/kg/min; P < .05) along with levels of butyrate-producing intestinal microbiota. Intestinal microbiota might be developed as therapeutic agents to increase insulin sensitivity in humans; www.trialregister.nl; registered at the Dutch Trial Register (NTR1776).


Assuntos
Glicemia/metabolismo , Fezes/microbiologia , Resistência à Insulina , Intestino Delgado/microbiologia , Síndrome Metabólica/terapia , Metagenoma , Adulto , Alcaligenes faecalis , Bacteroidetes , Índice de Massa Corporal , Clostridium , Escherichia coli , Eubacterium , Ácidos Graxos Voláteis/metabolismo , Fezes/química , Humanos , Masculino , Síndrome Metabólica/sangue , Pessoa de Meia-Idade , Oxalobacter formigenes , Estatísticas não Paramétricas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...