Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-39175183

RESUMO

OBJECTIVES: Food protein-induced enterocolitis syndrome (FPIES) is a severe type of non-IgE (immunoglobulin E)-mediated (NIM) food allergy, with cow's milk (CM) being the most common offending food. The relationship between the gut microbiota and its metabolites with the inflammatory process in infants with CM FPIES is unknown, although evidence suggests a microbial dysbiosis in NIM patients. This study was performed to contribute to the knowledge of the interaction between the gut microbiota and its derived metabolites with the local immune system in feces of infants with CM FPIES at diagnosis. METHODS: Twelve infants with CM FPIES and a matched healthy control group were recruited and the gut microbiota was investigated by 16S amplicon and shotgun sequencing. Fatty acids (FAs) were measured by gas chromatography, while immune factors were determined by enzyme-linked immunosorbent assay and Luminex technology. RESULTS: A specific pattern of microbiota in the gut of CM FPIES patients was found, characterized by a high abundance of enterobacteria. Also, an intense excretion of FAs in the feces of these infants was observed. Furthermore, correlations were found between fecal bifidobacteria and immune factors. CONCLUSION: These fecal determinations may be useful to gain insight into the pathophysiology of this syndrome and should be taken in consideration for future studies of FPIES patients.

2.
J Pediatr Gastroenterol Nutr ; 78(4): 836-845, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38344848

RESUMO

OBJECTIVE: Analyze fecal and blood samples at point of diagnosis in IgE mediated cow's milk protein allergy (CMPA) and non-IgE mediated (NIM)-CMPA patients to look for potential new biomarkers. PATIENTS AND METHODS: Fourteen patients with IgE mediated CMPA and 13 with NIM-CMPA were recruited in three hospitals in the north of Spain, and were compared with 25 infants from a control group of the same age range. To characterize intestinal microbiota, 16S rDNA gene and internal transcribed spacer amplicons of bifidobacteria were sequenced with Illumina technology. Fatty acids were analyzed by gas chromatography, meanwhile intestinal inflammation markers were quantified by enzyme-linked immunosorbent assay and a multiplex system. Immunological analysis of blood was performed by flow cytometry. RESULTS: The fecal results obtained in the NIM-CMPA group stand out. Among them, a significant reduction in the abundance of Bifidobacteriaceae and Bifidobacterium sequences with respect to controls was observed. Bifidobacterial species were also different, highlighting the lower abundance of Bifidobacterium breve sequences. Fecal calprotectin levels were found to be significantly elevated in relation to IgE mediated patients. Also, a higher excretion of IL-10 and a lower excretion of IL-1ra and platelet derived growth factor-BB was found in NIM-CMPA patients. CONCLUSIONS: The differential fecal parameters found in NIM-CMPA patients could be useful in the diagnosis of NIM food allergy to CM proteins.


Assuntos
Hipersensibilidade Alimentar , Microbioma Gastrointestinal , Hipersensibilidade a Leite , Lactente , Feminino , Animais , Humanos , Bovinos , Imunoglobulina E , Hipersensibilidade a Leite/diagnóstico , Proteínas do Leite
3.
Crit Rev Microbiol ; 49(5): 556-577, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35749433

RESUMO

Since its development in the 1960s, flow cytometry (FCM) was quickly revealed a powerful tool to analyse cell populations in medical studies, yet, for many years, was almost exclusively used to analyse eukaryotic cells. Instrument and methodological limitations to distinguish genuine bacterial signals from the background, among other limitations, have hampered FCM applications in bacteriology. In recent years, thanks to the continuous development of FCM instruments and methods with a higher discriminatory capacity to detect low-size particles, FCM has emerged as an appealing technique to advance the study of microbes, with important applications in research, clinical and industrial settings. The capacity to rapidly enumerate and classify individual bacterial cells based on viability facilitates the monitoring of bacterial presence in foodstuffs or clinical samples, reducing the time needed to detect contamination or infectious processes. Besides, FCM has stood out as a valuable tool to advance the study of complex microbial communities, or microbiomes, that are very relevant in the context of human health, as well as to understand the interaction of bacterial and host cells. This review highlights current developments in, and future applications of, FCM in bacteriology, with a focus on those related to food and clinical microbiology.


Assuntos
Bacteriologia , Humanos , Citometria de Fluxo/métodos , Bactérias/genética , Microbiologia de Alimentos
4.
J Appl Microbiol ; 133(1): 212-229, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35238463

RESUMO

Traditionally, fermentation was used to preserve the shelf life of food. Currently, in addition to favouring food preservation, well standardized and controlled industrial processes are also aimed at improving the functional characteristics of the final product. In this regard, starter cultures have become an essential cornerstone of food production. The selection of robust microorganisms, well adapted to the food environment, has been followed by the development of microbial consortia that provide some functional characteristics, beyond their acidifying capacity, achieving safer, high-quality foods with improved nutritional and health-promoting properties. In addition to starters, adjunct cultures and probiotics, which normally do not have a relevant role in fermentation, are added to the food in order to provide some beneficial characteristics. This review focuses on highlighting the functional characteristics of food starters, as well as adjunct and probiotic cultures (mainly lactic acid bacteria and bifidobacteria), with a specific focus on the synthesis of metabolites for preservation and safety aspects (e.g. bacteriocins), organoleptic properties (e.g. exopolysaccharides), nutritional (e.g. vitamins) and health improvement (e.g. neuroactive molecules). Literature reporting the application of these functional cultures in the manufacture of foods, mainly those related to dairy production, such as cheeses and fermented milks, has also been updated.


Assuntos
Queijo , Probióticos , Queijo/microbiologia , Fermentação , Microbiologia de Alimentos , Conservação de Alimentos , Valor Nutritivo , Probióticos/análise
5.
Int J Mol Sci ; 23(13)2022 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-35806099

RESUMO

Inflammatory bowel disease is a chronic disorder including ulcerative colitis and Crohn's disease (CD). Gut dysbiosis is often associated with CD, and metagenomics allows a better understanding of the microbial communities involved. The objective of this study was to reconstruct in silico carbohydrate metabolic capabilities from metagenome-assembled genomes (MAGs) obtained from healthy and CD individuals. This computational method was developed as a mean to aid rationally designed prebiotic interventions to rebalance CD dysbiosis, with a focus on metabolism of emergent prebiotics derived from arabinoxylan and pectin. Up to 1196 and 1577 MAGs were recovered from CD and healthy people, respectively. MAGs of Akkermansia muciniphila, Barnesiella viscericola DSM 18177 and Paraprevotella xylaniphila YIT 11841 showed a wide range of unique and specific enzymes acting on arabinoxylan and pectin. These glycosidases were also found in MAGs recovered from CD patients. Interestingly, these arabinoxylan and pectin degraders are predicted to exhibit metabolic interactions with other gut microbes reduced in CD. Thus, administration of arabinoxylan and pectin may ameliorate dysbiosis in CD by promoting species with key metabolic functions, capable of cross-feeding other beneficial species. These computational methods may be of special interest for the rational design of prebiotic ingredients targeting at CD.


Assuntos
Doença de Crohn , Microbiota , Doença de Crohn/tratamento farmacológico , Disbiose , Humanos , Pectinas , Xilanos
6.
Environ Microbiol ; 23(6): 3294-3305, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33973321

RESUMO

Whole metagenomic shotgun (WMS) sequencing has dramatically enhanced our ability to study microbial genomics. The possibility to unveil the genetic makeup of bacteria that cannot be easily isolated has significantly expanded our microbiological horizon. Here, we report an approach aimed at uncovering novel bacterial species by the use of targeted WMS sequencing. Employing in silico data retrieved from metabolic modelling to formulate a chemically defined medium (CDM), we were able to isolate and subsequently sequence the genomes of six putative novel species of bacteria from the gut of non-human primates.


Assuntos
Microbioma Gastrointestinal , Animais , Bactérias/genética , Microbioma Gastrointestinal/genética , Genoma Bacteriano/genética , Metagenoma , Metagenômica
7.
Artigo em Inglês | MEDLINE | ID: mdl-34398726

RESUMO

A strictly anaerobic, resistant starch-degrading, bile-tolerant, autolytic strain, IPLA60002T, belonging to the family Ruminococcaceae, was isolated from a human bile sample of a liver donor without hepatobiliary disease. Cells were Gram-stain-positive cocci, and 16S rRNA gene and whole genome analyses showed that Ruminococcus bromii was the phylogenetically closest related species to the novel strain IPLA60002T, though with average nucleotide identity values below 90 %. Biochemically, the new isolate has metabolic features similar to those described previously for gut R. bromii strains, including the ability to degrade a range of different starches. The new isolate, however, produces lactate and shows distinct resistance to the presence of bile salts. Additionally, the novel bile isolate displays an autolytic phenotype after growing in different media. Strain IPLA60002T is phylogenetically distinct from other species within the genus Ruminococcus. Therefore, we propose on the basis of phylogenetic, genomic and metabolic data that the novel IPLA60002T strain isolated from human bile be given the name Ruminococcoides bili gen. nov., sp. nov., within the new proposed genus Ruminococcoides and the family Ruminococcaceae. Strain IPLA60002T (=DSM 110008T=LMG 31505T) is proposed as the type strain of Ruminococcoides bili.


Assuntos
Bile/microbiologia , Filogenia , Ruminococcus/classificação , Técnicas de Tipagem Bacteriana , Composição de Bases , DNA Bacteriano/genética , Ácidos Graxos/química , Humanos , RNA Ribossômico 16S/genética , Ruminococcus/isolamento & purificação , Análise de Sequência de DNA
8.
Int Microbiol ; 24(4): 593-605, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34686940

RESUMO

The study of the food microbiome has gained considerable interest in recent years, mainly due to the wide range of applications that can be derived from the analysis of metagenomes. Among these applications, it is worth mentioning the possibility of using metagenomic analyses to determine food authenticity, to assess the microbiological safety of foods thanks to the detection and tracking of pathogens, antibiotic resistance genes and other undesirable traits, as well to identify the microorganisms responsible for food processing defects. Metataxonomics and metagenomics are currently the gold standard methodologies to explore the full potential of metagenomes in the food industry. However, there are still a number of challenges that must be solved in order to implement these methods routinely in food chain monitoring, and for the regulatory agencies to take them into account in their opinions. These challenges include the difficulties of analysing foods and food-related environments with a low microbial load, the lack of validated bioinformatics pipelines adapted to food microbiomes and the difficulty of assessing the viability of the detected microorganisms. This review summarizes the methods of microbiome analysis that have been used, so far, in foods and food-related environments, with a specific focus on those involving Next-Generation Sequencing technologies.


Assuntos
Metagenômica , Microbiota , Resistência Microbiana a Medicamentos , Indústria Alimentícia , Metagenoma
9.
Int J Mol Sci ; 22(3)2021 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-33498321

RESUMO

The gut microbiota has emerged as a key factor in the pathogenesis of intestinal viruses, including enteroviruses, noroviruses and rotaviruses (RVs), where stimulatory and inhibitory effects on infectivity have been reported. With the aim of determining whether members of the microbiota interact with RVs during infection, a combination of anti-RV antibody labeling, fluorescence-activated cell sorting and 16S rRNA amplicon sequencing was used to characterize the interaction between specific bacteria and RV in stool samples of children suffering from diarrhea produced by G1P[8] RV. The genera Ruminococcus and Oxalobacter were identified as RV binders in stools, displaying enrichments between 4.8- and 5.4-fold compared to samples nonlabeled with anti-RV antibodies. In vitro binding of the G1P[8] Wa human RV strain to two Ruminococcus gauvreauii human isolates was confirmed by fluorescence microscopy. Analysis in R. gauvreauii with antibodies directed to several histo-blood group antigens (HBGAs) indicated that these bacteria express HBGA-like substances on their surfaces, which can be the target for RV binding. Furthermore, in vitro infection of the Wa strain in differentiated Caco-2 cells was significantly reduced by incubation with R. gauvreauii. These data, together with previous findings showing a negative correlation between Ruminococcus levels and antibody titers to RV in healthy individuals, suggest a pivotal interaction between this bacterial group and human RV. These results reveal likely mechanisms of how specific bacterial taxa of the intestinal microbiota could negatively affect RV infection and open new possibilities for antiviral strategies.


Assuntos
Microbioma Gastrointestinal , Infecções por Rotavirus/microbiologia , Rotavirus/metabolismo , Ruminococcus/metabolismo , Proteínas de Bactérias/metabolismo , Células CACO-2 , Pré-Escolar , Humanos , Intestinos/microbiologia , Intestinos/virologia , Ligação Proteica , Rotavirus/patogenicidade , Infecções por Rotavirus/virologia , Ruminococcus/patogenicidade
10.
Environ Microbiol ; 22(8): 3375-3393, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32515117

RESUMO

In recent years, bifidobacterial populations in the gut of various monkey species have been assessed in several ecological surveys, unveiling a diverse, yet unexplored ecosystem harbouring novel species. In the current study, we investigated the species distribution of bifidobacteria present in 23 different species of primates, including human samples, by means of 16S rRNA microbial profiling and internal transcribed spacer bifidobacterial profiling. Based on the observed bifidobacterial-host co-phylogeny, we found a statistically significant correlation between the Hominidae family and particular bifidobacterial species isolated from humans, indicating phylosymbiosis between these lineages. Furthermore, phylogenetic and glycobiome analyses, based on 40 bifidobacterial species isolated from primates, revealed that members of the Bifidobacterium tissieri phylogenetic group, which are typical gut inhabitants of members of the Cebidae family, descend from an ancient ancestor with respect to other bifidobacterial taxa isolated from primates.


Assuntos
Bifidobacterium/classificação , Bifidobacterium/isolamento & purificação , Microbioma Gastrointestinal/genética , Primatas/microbiologia , Animais , Bifidobacterium/genética , Evolução Biológica , Ecossistema , Humanos , Filogenia , RNA Ribossômico 16S/genética
11.
J Neuroinflammation ; 17(1): 363, 2020 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-33246492

RESUMO

BACKGROUND: Multiple sclerosis (MS) is a chronic demyelinating autoimmune disease affecting the CNS. Recent studies have indicated that intestinal alterations play key pathogenic roles in the development of autoimmune diseases, including MS. The triterpene oleanolic acid (OA), due to its anti-inflammatory properties, has shown to beneficially influence the severity of the experimental autoimmune encephalomyelitis (EAE), a preclinical model of MS. We herein investigate EAE-associated gut intestinal dysfunction and the effect of OA treatment. METHODS: Mice with MOG35-55-induced EAE were treated with OA or vehicle from immunization day and were daily analyzed for clinical deficit. We performed molecular and histological analysis in serum and intestinal tissues to measure oxidative and inflammatory responses. We used Caco-2 and HT29-MTX-E12 cells to elucidate OA in vitro effects. RESULTS: We found that OA protected from EAE-induced changes in intestinal permeability and preserved the mucin-containing goblet cells along the intestinal tract. Serum levels of the markers for intestinal barrier damage iFABP and monocyte activation sCD14 were consistently and significantly reduced in OA-treated EAE mice. Beneficial OA effects also included a decrease of pro-inflammatory mediators both in serum and colonic tissue of treated-EAE mice. Moreover, the levels of some immunoregulatory cytokines, the neurotrophic factor GDNF, and the gastrointestinal hormone motilin were preserved in OA-treated EAE mice. Regarding oxidative stress, OA treatment prevented lipid peroxidation and superoxide anion accumulation in intestinal tissue, while inducing the expression of the ROS scavenger Sestrin-3. Furthermore, short-chain fatty acids (SCFA) quantification in the cecal content showed that OA reduced the high iso-valeric acid concentrations detected in EAE-mice. Lastly, using in vitro cell models which mimic the intestinal epithelium, we verified that OA protected against intestinal barrier dysfunction induced by injurious agents produced in both EAE and MS. CONCLUSION: These findings reveal that OA ameliorates the gut dysfunction found in EAE mice. OA normalizes the levels of gut mucosal dysfunction markers, as well as the pro- and anti-inflammatory immune bias during EAE, thus reinforcing the idea that OA is a beneficial compound for treating EAE and suggesting that OA may be an interesting candidate to be explored for the treatment of human MS.


Assuntos
Encefalomielite Autoimune Experimental/patologia , Mucosa Intestinal/efeitos dos fármacos , Ácido Oleanólico/farmacologia , Animais , Células CACO-2 , Feminino , Células HT29 , Humanos , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/patologia , Estresse Oxidativo/efeitos dos fármacos , Permeabilidade/efeitos dos fármacos
12.
Anaerobe ; 56: 98-101, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30794874

RESUMO

The toll-like receptors involved in recognition of the exopolysaccharide produced by two isogenic, ropy and non-ropy, Bifidobacterium animalis subsp. lactis strains were investigated. Both strains interact with human embryonic kidney (HEK)-293 cells via TLR2, whereas purified EPSs specifically stimulate TLR4 regardless their molar mass.


Assuntos
Bifidobacterium animalis/metabolismo , Células Epiteliais/metabolismo , Polissacarídeos Bacterianos/metabolismo , Receptor 4 Toll-Like/metabolismo , Linhagem Celular , Humanos , Ligação Proteica
13.
Appl Environ Microbiol ; 84(10)2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29500262

RESUMO

Bifidobacteria are mutualistic intestinal bacteria, and their presence in the human gut has been associated with health-promoting activities. The presence of antibiotic resistance genes in this genus is controversial, since, although bifidobacteria are nonpathogenic microorganisms, they could serve as reservoirs of resistance determinants for intestinal pathogens. However, until now, few antibiotic resistance determinants have been functionally characterized in this genus. In this work, we show that Bifidobacterium breve CECT7263 displays atypical resistance to erythromycin and clindamycin. In order to delimit the genomic region responsible for the observed resistance phenotype, a library of genomic DNA was constructed and a fragment of 5.8 kb containing a gene homologous to rRNA methylase genes was able to confer erythromycin resistance in Escherichia coli This genomic region seems to be very uncommon, and homologs of the gene have been detected in only one strain of Bifidobacterium longum and two other strains of B. breve In this context, analysis of shotgun metagenomics data sets revealed that the gene is also uncommon in the microbiomes of adults and infants. The structural gene and its upstream region were cloned into a B. breve-sensitive strain, which became resistant after acquiring the genetic material. In vitro conjugation experiments did not allow us to detect gene transfer to other recipients. Nevertheless, prediction of genes potentially acquired through horizontal gene transfer events revealed that the gene is located in a putative genomic island.IMPORTANCEBifidobacterium breve is a very common human intestinal bacterium. Often described as a pioneer microorganism in the establishment of early-life intestinal microbiota, its presence has been associated with several beneficial effects for the host, including immune stimulation and protection against infections. Therefore, some strains of this species are considered probiotics. In relation to this, because probiotic bacteria are used for human and animal consumption, one of the safety concerns over these bacteria is the presence of antibiotic resistance genes, since the human gut is a densely populated habitat that could favor the transfer of genetic material to potential pathogens. In this study, we analyzed the genetic basis responsible for the erythromycin and clindamycin resistance phenotype of B. breve CECT7263. We were able to identify and characterize a novel gene homologous to rRNA methylase genes which confers erythromycin and clindamycin resistance. This gene seems to be very uncommon in other bifidobacteria and in the gut microbiomes of both adults and infants. Even though conjugation experiments showed the absence of transferability under in vitro conditions, it has been predicted to be located in a putative genomic island recently acquired by specific bifidobacterial strains.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Bifidobacterium breve/efeitos dos fármacos , Bifidobacterium breve/enzimologia , Clindamicina/farmacologia , Eritromicina/farmacologia , Metiltransferases/metabolismo , Proteínas de Bactérias/genética , Bifidobacterium breve/genética , Farmacorresistência Bacteriana , Microbioma Gastrointestinal , Transferência Genética Horizontal , Humanos , Intestinos/microbiologia , Metiltransferases/genética , Filogenia
14.
Eur J Nutr ; 57(2): 487-497, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27744545

RESUMO

PURPOSE: Short-chain fatty acids (SCFAs) formation by intestinal bacteria is regulated by many different factors, among which dietary fibre is currently receiving most attention. However, since fibre-rich foods are usually good dietary sources of phenolic compounds, which are also known to affect the microbiota, authors hypothesize that the regular intake of these bioactive compounds could be associated with a modulation of faecal SCFA production by the intestinal microbiota. METHODS: In this work, food intake was recorded by means of a validated Food Frequency Questionnaire. Fibres were determined using Marlett food composition tables, and phenolic compounds were obtained from Phenol-Explorer Database. Analysis of SCFA was performed by gas chromatography-flame ionization/mass spectrometry and quantification of microbial populations in faeces by quantitative PCR. RESULTS: Klason lignin and its food contributors, as predictors of faecal butyrate production, were directly associated with Bacteroides and Bifidobacterium levels, as well as lignans with Bacteroides. Also, anthocyanidins, provided by strawberries, were associated with faecal propionate and inversely related to Lactobacillus group. CONCLUSIONS: These results support the hypothesis we put forward regarding the association between some vegetable foods (strawberries, pasta, lentils, lettuce and olive oil) and faecal SCFA. More studies are needed in order to elucidate whether these associations have been mediated by the bacterial modulatory effect of the bioactive compounds, anthocyanins, lignans or Klason lignin, present in foodstuffs.


Assuntos
Bacteroides/metabolismo , Bifidobacterium/metabolismo , Dieta Saudável , Fibras na Dieta/uso terapêutico , Disbiose/prevenção & controle , Microbioma Gastrointestinal , Cooperação do Paciente , Adulto , Idoso , Idoso de 80 Anos ou mais , Bacteroides/classificação , Bacteroides/crescimento & desenvolvimento , Bacteroides/isolamento & purificação , Bifidobacterium/classificação , Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/isolamento & purificação , Estudos Transversais , Dieta/efeitos adversos , Dieta/etnologia , Dieta Saudável/etnologia , Fibras na Dieta/metabolismo , Disbiose/etnologia , Disbiose/etiologia , Disbiose/microbiologia , Ácidos Graxos Voláteis/análise , Ácidos Graxos Voláteis/metabolismo , Fezes/química , Fezes/microbiologia , Feminino , Fermentação , Humanos , Masculino , Pessoa de Meia-Idade , Tipagem Molecular , Avaliação Nutricional , Inquéritos Nutricionais , Cooperação do Paciente/etnologia , Espanha , Adulto Jovem
16.
Environ Microbiol ; 19(4): 1536-1551, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28251782

RESUMO

Recent evidence has disclosed a connection between gut microbial glycosidase activity and adiposity in obese. Here, we measured microbial α-glucosidase and ß-galactosidase activities and sorted fluorescently labeled ß-galactosidase containing (ßGAL) microorganisms in faecal samples of eight lean and thirteen obese adolescents that followed a controlled calorie restriction program during one year. ß-galactosidase is a highly distributed functional trait, mainly expressed by members of Blautia, Bacteroides, Alcaligenes, Acinetobacter and Propionibacterium. Only long-term calorie restriction induced clear changes in the microbiota of obese adolescents. Long-term calorie restriction induced significant shifts in total and ßGAL gut microbiota, reducing the Firmicutes:Bacteroidetes ratio and enhancing the growth of beneficial microorganisms such as Bacteroides, Roseburia, Faecalibacterium and Clostridium XIVa. Moreover, the structure and composition of ßGAL community in obese after long-term calorie restriction was highly similar to that of lean adolescents. However, despite this high compositional similarity, microbial metabolic performance was different, split in two metabolic states at a body mass index value of 25. Our study shows that calorie restriction is a strong environmental force reshaping gut microbiota though its metabolic performance is linked to host's adiposity, suggesting that functional redundancy and metabolic plasticity are fundamental properties of gut microbial ecosystem.


Assuntos
Restrição Calórica , Microbioma Gastrointestinal , Obesidade/microbiologia , Adolescente , Bacteroides , Bacteroidetes , Clostridium , Fezes/microbiologia , Humanos
17.
Curr Opin Rheumatol ; 28(5): 515-22, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27466725

RESUMO

PURPOSE OF REVIEW: Recent discoveries relay commensal gut microbiota as a relevant factor in the maintenance of intestinal homeostasis. RECENT FINDINGS: Alterations in the composition of the intestinal microbiota have been reported in patients with systemic lupus erythematosus and many other inflammatory and autoimmune conditions. However, the mechanisms by which the intestinal microbiota can influence systemic immunity in these situations remain to be elucidated. The inappropriate immune responses of patients with systemic lupus erythematosus could originate a breakdown of tolerance towards the microbiota, leading to the expansion and/or contraction of specific bacterial groups that may culminate in a dysbiotic state. Conversely, an altered composition of the intestinal microbiome in genetically predisposed individuals could influence systemic immunity by several mechanisms, leading to a breakdown of tolerance to self-antigens. Moreover, humoral immune responses can be affected by specific bacterial groups in these individuals. SUMMARY: Recent findings support an important role for the crosstalk between bacteria and immune cells to maintain an intestinal homeostasis crucial to sustain tolerance toward self-antigens and intestinal microbiota.


Assuntos
Anticorpos Antinucleares/imunologia , Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Intestinos/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Tolerância a Antígenos Próprios/imunologia , Doenças Autoimunes/imunologia , Células Dendríticas/imunologia , Homeostase , Humanos , Tolerância Imunológica/imunologia , Intestinos/microbiologia , Linfócitos/imunologia , Macrófagos/imunologia , Células Mieloides/imunologia
18.
Proc Natl Acad Sci U S A ; 110(27): 11151-6, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23776216

RESUMO

Bifidobacteria represent one of the dominant groups of microorganisms colonizing the human infant intestine. Commensal bacteria that interact with a eukaryotic host are believed to express adhesive molecules on their cell surface that bind to specific host cell receptors or soluble macromolecules. Whole-genome transcription profiling of Bifidobacterium bifidum PRL2010, a strain isolated from infant stool, revealed a small number of commonly expressed extracellular proteins, among which were genes that specify sortase-dependent pili. Expression of the coding sequences of these B. bifidum PRL2010 appendages in nonpiliated Lactococcus lactis enhanced adherence to human enterocytes through extracellular matrix protein and bacterial aggregation. Furthermore, such piliated L. lactis cells evoked a higher TNF-α response during murine colonization compared with their nonpiliated parent, suggesting that bifidobacterial sortase-dependent pili not only contribute to adherence but also display immunomodulatory activity.


Assuntos
Bifidobacterium/fisiologia , Fímbrias Bacterianas/fisiologia , Aminoaciltransferases/genética , Aminoaciltransferases/metabolismo , Animais , Aderência Bacteriana/genética , Aderência Bacteriana/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Bifidobacterium/genética , Bifidobacterium/imunologia , Linhagem Celular , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Citocinas/biossíntese , Feminino , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/imunologia , Genes Bacterianos , Humanos , Lactente , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Lactococcus lactis/genética , Lactococcus lactis/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Probióticos , Transcriptoma/imunologia
19.
Int J Mol Sci ; 17(5)2016 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-27136545

RESUMO

BACKGROUND: The microbial colonization of the neonatal gut provides a critical stimulus for normal maturation and development. This process of early microbiota establishment, known to be affected by several factors, constitutes an important determinant for later health. METHODS: We studied the establishment of the microbiota in preterm and full-term infants and the impact of perinatal antibiotics upon this process in premature babies. To this end, 16S rRNA gene sequence-based microbiota assessment was performed at phylum level and functional inference analyses were conducted. Moreover, the levels of the main intestinal microbial metabolites, the short-chain fatty acids (SCFA) acetate, propionate and butyrate, were measured by Gas-Chromatography Flame ionization/Mass spectrometry detection. RESULTS: Prematurity affects microbiota composition at phylum level, leading to increases of Proteobacteria and reduction of other intestinal microorganisms. Perinatal antibiotic use further affected the microbiota of the preterm infant. These changes involved a concomitant alteration in the levels of intestinal SCFA. Moreover, functional inference analyses allowed for identifying metabolic pathways potentially affected by prematurity and perinatal antibiotics use. CONCLUSION: A deficiency or delay in the establishment of normal microbiota function seems to be present in preterm infants. Perinatal antibiotic use, such as intrapartum prophylaxis, affected the early life microbiota establishment in preterm newborns, which may have consequences for later health.


Assuntos
Antibacterianos/farmacologia , Intestinos/microbiologia , Microbiota/efeitos dos fármacos , Acetatos/análise , Aleitamento Materno , Butiratos/análise , Ácidos Graxos Voláteis/análise , Fezes/química , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Propionatos/análise
20.
Environ Microbiol ; 17(7): 2515-31, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25523018

RESUMO

Bifidobacteria are bacterial gut commensals of mammals, birds and social insects that are perceived to influence the metabolism/physiology of their host. In this context, members of the Bifidobacterium bifidum species are believed to significantly contribute to the overall microbiota of the human gut at infant stage. However, the molecular reasons for their adaptation to this environment are poorly understood. In this study, we analysed the pan-genome of B. bifidum species by decoding genomes of 15 B. bifidum strains, which highlighted the existence of a conserved gene uniquely present in this bifidobacterial taxon, underscoring a nutrient acquisition strategy that targets host-derived glycans, such as those present in mucin. Growth experiments and corresponding transcriptomic analyses confirmed the in silico data and supported these intriguing and unique host glycan-specific saccharolytic features. The ubiquity of the genetic features of B. bifidum for the breakdown of host glycans was confirmed by interrogating metagenomic datasets, thereby supporting the notion that metabolic access to host-derived glycans is a potent evolutionary force that has shaped B. bifidum genomes and consequently the ecology of the infant intestinal microbiota.


Assuntos
Bifidobacterium/genética , Bifidobacterium/metabolismo , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Polissacarídeos/metabolismo , Animais , Bifidobacterium/crescimento & desenvolvimento , Evolução Biológica , Genoma Bacteriano , Humanos , Lactente , Recém-Nascido , Dados de Sequência Molecular , Mucinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA