Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Appl Environ Microbiol ; 86(23)2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-32948523

RESUMEN

In recent years, various studies have demonstrated that the gut microbiota influences host metabolism. However, these studies were focused primarily on a single or a limited range of host species, thus preventing a full exploration of possible taxonomic and functional adaptations by gut microbiota members as a result of host-microbe coevolution events. In the current study, the microbial taxonomic profiles of 250 fecal samples, corresponding to 77 host species that cover the mammalian branch of the tree of life, were reconstructed by 16S rRNA gene-based sequence analysis. Moreover, shotgun metagenomics was employed to investigate the metabolic potential of the fecal microbiomes of 24 mammals, and subsequent statistical analyses were performed to assess the impact of host diet and corresponding physiology of the digestive system on gut microbiota composition and functionality. Functional data were confirmed and extended through metatranscriptome assessment of gut microbial populations of eight animals, thus providing insights into the transcriptional response of gut microbiota to specific dietary lifestyles. Therefore, the analyses performed in this study support the notion that the metabolic features of the mammalian gut microbiota have adapted to maximize energy extraction from the host's diet.IMPORTANCE Diet and host physiology have been recognized as main factors affecting both taxonomic composition and functional features of the mammalian gut microbiota. However, very few studies have investigated the bacterial biodiversity of mammals by using large sample numbers that correspond to multiple mammalian species, thus resulting in an incomplete understanding of the functional aspects of their microbiome. Therefore, we investigated the bacterial taxonomic composition of 250 fecal samples belonging to 77 host species distributed along the tree of life in order to assess how diet and host physiology impact the intestinal microbial community by selecting specific microbial players. Conversely, the application of shotgun metagenomics and metatranscriptomics approaches to a group of selected fecal samples allowed us to shed light on both metabolic features and transcriptional responses of the intestinal bacterial community based on different diets.


Asunto(s)
Bacterias/aislamiento & purificación , Dieta/veterinaria , Heces/microbiología , Microbioma Gastrointestinal , Mamíferos/microbiología , Mamíferos/fisiología , Animales , Bacterias/clasificación , Perfilación de la Expresión Génica/veterinaria , Metagenómica , ARN Bacteriano/análisis , ARN Ribosómico 16S/análisis , Especificidad de la Especie
2.
mSystems ; 5(4)2020 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-32723792

RESUMEN

16S small-subunit (SSU) rRNA gene-based bacterial profiling is the gold standard for cost-effective taxonomic reconstruction of complex bacterial populations down to the genus level. However, it has been proven ineffective in clinical and research settings requiring higher taxonomic resolution. We therefore developed a bacterial profiling method based on the internal transcribed spacer (ITS) region employing optimized primers and a comprehensive ITS database for accurate cataloguing of bacterial communities at (sub)species resolution. Performance of the microbial ITS profiling pipeline was tested through analysis of host-associated, food, and environmental matrices, while its efficacy in clinical settings was assessed through analysis of mucosal biopsy specimens of colorectal cancer, leading to the identification of putative novel biomarkers. The data collected indicate that the proposed pipeline represents a major step forward in cost-effective identification and screening of microbial biomarkers at (sub)species level, with relevant impact in research, industrial, and clinical settings.IMPORTANCE We developed a novel method for accurate cataloguing of bacterial communities at (sub)species level involving amplification of the internal transcribed spacer (ITS) region through optimized primers, followed by next-generation sequencing and taxonomic classification of amplicons by means of a comprehensive database of bacterial ITS sequences. Host-associated, food, and environmental matrices were employed to test the performance of the microbial ITS profiling pipeline. Moreover, mucosal biopsy samples from colorectal cancer patients were analyzed to demonstrate the scientific relevance of this profiling approach in a clinical setting through identification of putative novel biomarkers. The results indicate that the ITS-based profiling pipeline proposed here represents a key metagenomic tool with major relevance for research, industrial, and clinical settings.

3.
FEMS Microbiol Ecol ; 95(9)2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31344227

RESUMEN

Domestication is the process by which anthropogenic forces shape lifestyle and behavior of wild species to accommodate human needs. The impact of domestication on animal physiology and behavior has been extensively studied, whereas its effect on the gut microbiota is still largely unexplored. For this reason, 16S rRNA gene-based and internal transcribed spacer-mediated bifidobacterial profiling, together with shotgun metagenomics, was employed to investigate the taxonomic composition and metabolic repertoire of 146 mammalian fecal samples, corresponding to 12 domesticated-feral dyads. Our results revealed that changes induced by domestication have extensively shaped the taxonomic composition of the mammalian gut microbiota. In this context, the selection of microbial taxa linked to a more efficient feed conversion into body mass and putative horizontal transmission of certain bacterial genera from humans were observed in the fecal microbiota of domesticated animals when compared to their feral relatives and to humans. In addition, profiling of the metabolic arsenal through metagenomics highlighted extensive functional adaptation of the fecal microbial community of domesticated mammals to changes induced by domestication. Remarkably, domesticated animals showed, when compared to their feral relatives, increased abundance of specific glycosyl hydrolases, possibly due to the higher intake of complex plant carbohydrates typical of commercial animal feeds.


Asunto(s)
Bacterias/aislamiento & purificación , Microbioma Gastrointestinal , Mamíferos/microbiología , Alimentación Animal/análisis , Animales , Bacterias/clasificación , Bacterias/genética , Domesticación , Heces/microbiología , Humanos , Mamíferos/crecimiento & desarrollo , Metagenómica , Microbiota , ARN Ribosómico 16S/genética
4.
Microbiome ; 7(1): 100, 2019 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-31272480

RESUMEN

BACKGROUND: The microbial populations of the human intestinal tract and their relationship to specific diseases have been extensively studied during the last decade. However, the characterization of the human bile microbiota as a whole has been hampered by difficulties in accessing biological samples and the lack of adequate methodologies to assess molecular studies. Although a few reports have described the biliary microbiota in some hepatobiliary diseases, the bile microbiota of healthy individuals has not been described. With this in mind, the goal of the present study was to generate fundamental knowledge on the composition and activity of the human bile microbiota, as well as establishing its potential relationship with human bile-related disorders. RESULTS: Human bile samples from the gallbladder of individuals from a control group, without any record of hepatobiliary disorder, were obtained from liver donors during liver transplantation surgery. A bile DNA extraction method was optimized together with a quantitative PCR (qPCR) assay for determining the bacterial load. This allows the selection of samples to perform functional metagenomic analysis. Bile samples from the gallbladder of individuals suffering from lithiasis were collected during gallbladder resection and the microbial profiles assessed, using a 16S rRNA gene-based sequencing analysis, and compared with those of the control group. Additionally, the metabolic profile of the samples was analyzed by nuclear magnetic resonance (NMR). We detected, for the first time, bacterial communities in gallbladder samples of individuals without any hepatobiliary pathology. In the biliary microecosystem, the main bacterial phyla were represented by Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria. Significant differences in the relative abundance of different taxa of both groups were found. Sequences belonging to the family Propionibacteriaceae were more abundant in bile samples from control subjects; meanwhile, in patients with cholelithiasis members of the families Bacteroidaceae, Prevotellaceae, Porphyromonadaceae, and Veillonellaceae were more frequently detected. Furthermore, the metabolomics analysis showed that the two study groups have different metabolic profiles. CONCLUSIONS: Our results indicate that the gallbladder of human individuals, without diagnosed hepatobiliary pathology, harbors a microbial ecosystem that is described for the first time in this study. Its bacterial representatives and metabolites are different from those detected in people suffering from cholelithiasis. In this regard, since liver donors have been subjected to the specific conditions of the hospital's intensive care unit, including an antibiotic treatment, we must be cautious in stating that their bile samples contain a physiologically normal biliary microbiome. In any case, our results open up new possibilities to discover bacterial functions in a microbial ecosystem that has not previously been explored.


Asunto(s)
Bilis/metabolismo , Bilis/microbiología , Vesícula Biliar/microbiología , Vesícula Biliar/fisiología , Microbiota , Adulto , Anciano , Bacterias/clasificación , Femenino , Humanos , Litiasis/microbiología , Masculino , Metabolómica , Metagenoma , Persona de Mediana Edad , ARN Ribosómico 16S/genética , ARN Ribosómico 18S/genética
5.
Environ Microbiol ; 21(10): 3683-3695, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31172651

RESUMEN

Bifidobacterium bifidum is reported to be among the first colonizers of the newborn's gastrointestinal tract due to its ability to metabolize human milk oligosaccharides (HMOs). In order to investigate biological features that allow this bifidobacterial species to colonize a newborn, bifidobacterial internally transcribed spacer profiling of stool samples of 50 mother-infant dyads, as well as corresponding breastmilk samples, was performed. Hierarchical clustering based on bifidobacterial population profiles found in infant faecal samples revealed the presence of four bifidobacterial clusters or the so-called bifidotypes. Bifidobacterium bifidum was shown to be a key member among bifidotypes, in which its presence correlate with several different bifidobacterial species retrieved in infant faecal samples. For this reason, we investigated cross-feeding behaviour facilitated by B. bifidum on a bioreactor model using human milk as growth substrate. Transcriptional profiles of this strain were evaluated when grown on nine specific glycans typically constituting HMOs. Remarkably, these analyses suggest extensive co-evolution with the host and other bifidobacterial species in terms of resource provision and sharing, respectively, activities that appear to support a bifidobacteria-dominant microbiome.


Asunto(s)
Bifidobacterium bifidum/fisiología , Coevolución Biológica , Microbioma Gastrointestinal , Adolescente , Adulto , Reactores Biológicos , Heces/microbiología , Femenino , Humanos , Recién Nacido , Leche Humana/microbiología , Oligosacáridos/metabolismo , Polisacáridos/metabolismo , Adulto Joven
6.
Nat Commun ; 10(1): 1286, 2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30894548

RESUMEN

The abilities of certain microorganisms to be transferred across the food production chain, persist in the final product and, potentially, colonize the human gut are poorly understood. Here, we provide strain-level evidence supporting that dairy cattle-associated bacteria can be transferred to the human gut via consumption of Parmesan cheese. We characterize the microbial communities in samples taken from five different locations across the Parmesan cheese production chain, confirming that the final product contains microorganisms derived from cattle gut, milk, and the nearby environment. In addition, we carry out a human pilot study showing that Bifidobacterium mongoliense strains from cheese can transiently colonize the human gut, a process that can be enhanced by cow milk consumption.


Asunto(s)
Queso/microbiología , ADN Bacteriano/genética , Microbioma Gastrointestinal/genética , Leche/microbiología , Filogenia , Animales , Bifidobacterium/clasificación , Bifidobacterium/genética , Bifidobacterium/aislamiento & purificación , Bovinos , Corynebacterium/clasificación , Corynebacterium/genética , Corynebacterium/aislamiento & purificación , Código de Barras del ADN Taxonómico , Heces/microbiología , Humanos , Lactobacillus delbrueckii/clasificación , Lactobacillus delbrueckii/genética , Lactobacillus delbrueckii/aislamiento & purificación , Proyectos Piloto , Prevotella ruminicola/clasificación , Prevotella ruminicola/genética , Prevotella ruminicola/aislamiento & purificación , ARN Ribosómico 16S/genética , Streptococcus thermophilus/clasificación , Streptococcus thermophilus/genética , Streptococcus thermophilus/aislamiento & purificación
7.
Int J Food Microbiol ; 294: 1-9, 2019 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-30711887

RESUMEN

Health promoting or probiotic bacteria are commonly incorporated into a variety of functional foods and drug formulations, due to their purported ability to confer benefit to host health. Despite the extensive commercial exploitation of probiotic formulations there are still major knowledge gaps regarding the precise molecular mechanism of action and corresponding genetic/genomic properties of probiotic bacteria. In the current study, we describe a metagenomic approach which allows determination of the composition of probiotic supplements through next-generation sequencing analyses based on rRNA-associated sequences to assess bacterial composition of the product combined with a shotgun metagenomics approach directed to decode the genome sequences of the probiotic strains for each product assayed. The here developed approach has been tested for 10 probiotic supplements, revealing inconsistencies between the identified probiotic strains and the declared strains as indicated by the producers. Furthermore, the decoded bacterial genome sequence of Bifidobacterium animalis subsp. lactis BB-12 from a 1995 frozen dried stock revealed genetic evidence for genome evolution and stability of this microorganism when compared with the re-constructed genome of the identical strain from a probiotic supplement of 2017.


Asunto(s)
Bacterias/clasificación , Bacterias/genética , Suplementos Dietéticos/microbiología , Microbiología de Alimentos/métodos , Genoma Bacteriano/genética , Metagenómica , Probióticos/análisis , Bifidobacterium animalis/clasificación , Bifidobacterium animalis/genética , ADN Ribosómico/genética
8.
Appl Environ Microbiol ; 85(7)2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30709821

RESUMEN

Bifidobacteria are members of the gut microbiota of animals, including mammals, birds, and social insects. In this study, we analyzed and determined the pangenome of Bifidobacterium animalis species, encompassing B. animalis subsp. animalis and the B. animalis subsp. lactis taxon, which is one of the most intensely exploited probiotic bifidobacterial species. In order to reveal differences within the B. animalis species, detailed comparative genomics and phylogenomics analyses were performed, indicating that these two subspecies recently arose through divergent evolutionary events. A subspecies-specific core genome was identified for both B. animalis subspecies, revealing the existence of subspecies-defining genes involved in carbohydrate metabolism. Notably, these in silico analyses coupled with carbohydrate profiling assays suggest genetic adaptations toward a distinct glycan milieu for each member of the B. animalis subspecies, resulting in a divergent evolutionary development of the two subspecies.IMPORTANCE The majority of characterized B. animalis strains have been isolated from human fecal samples. In order to explore genome variability within this species, we isolated 15 novel strains from the gastrointestinal tracts of different animals, including mammals and birds. The present study allowed us to reconstruct the pangenome of this taxon, including the genome contents of 56 B. animalis strains. Through careful assessment of subspecies-specific core genes of the B. animalis subsp. animalis/lactis taxon, we identified genes encoding enzymes involved in carbohydrate transport and metabolism, while unveiling specific gene acquisition and loss events that caused the evolutionary emergence of these two subspecies.


Asunto(s)
Bifidobacterium animalis/genética , Hibridación Genómica Comparativa , Evolución Molecular , Genes Bacterianos/genética , Filogenia , Animales , Bifidobacterium/genética , Bifidobacterium animalis/enzimología , Bifidobacterium animalis/metabolismo , Aves , Metabolismo de los Hidratos de Carbono , Carbohidratos , Heces/microbiología , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Variación Genética , Genoma Bacteriano/genética , Humanos , Mamíferos , Polisacáridos , Especificidad de la Especie
9.
Environ Microbiol ; 21(4): 1331-1343, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30680877

RESUMEN

Domestication of dogs from wolves is the oldest known example of ongoing animal selection, responsible for generating more than 300 dog breeds worldwide. In order to investigate the taxonomic and functional evolution of the canine gut microbiota, a multi-omics approach was applied to six wild wolves and 169 dog faecal samples, the latter encompassing 51 breeds, which fully covers currently known canine genetic biodiversity. Specifically, 16S rRNA gene and bifidobacterial Internally Transcribed Spacer (ITS) profiling were employed to reconstruct and then compare the canine core gut microbiota to those of wolves and humans, revealing that artificial selection and subsequent cohabitation of dogs with their owners influenced the microbial population of canine gut through loss and acquisition of specific bacterial taxa. Moreover, comparative analysis of the intestinal bacterial population of dogs fed on Bones and Raw Food (BARF) or commercial food (CF) diet, coupled with shotgun metagenomics, highlighted that both bacterial composition and metabolic repertoire of the canine gut microbiota have evolved to adapt to high-protein or high-carbohydrates intake. Altogether, these data indicate that artificial selection and domestication not only affected the canine genome, but also shaped extensively the bacterial population harboured by the canine gut.


Asunto(s)
Bacterias/clasificación , Bacterias/genética , Biodiversidad , Perros/microbiología , Microbioma Gastrointestinal/genética , Metagenoma/genética , Fenómenos Fisiológicos de la Nutrición , Animales , Bifidobacterium/genética , Heces/microbiología , Metagenómica , ARN Ribosómico 16S/genética , Lobos/microbiología
10.
Sci Rep ; 8(1): 13974, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30228361

RESUMEN

The human intestine retains a complex microbial ecosystem, which performs crucial functions that impact on host health. Several studies have indicated that intestinal dysbiosis may impact on the establishment of life-threatening intestinal diseases such as colorectal cancer. An adenomatous polyp is the result of abnormal tissue growth, which is benign but is considered to be associated with a high risk of developing colorectal cancer, based on its grade of dysplasia. Development of diagnostic tools that are based on surveying the gut microbiota and are aimed at early detection of colorectal cancer represent highly desirable target. For this purpose, we performed a pilot study in which we applied a metataxonomic analysis based on 16S rRNA gene sequencing approach to unveil the composition of microbial communities of intestinal polyps. Moreover, we performed a meta-analysis involving the reconstructed microbiota composition of adenomatous polyps and publicly available metagenomics datasets of colorectal cancer. These analyses allowed the identification of microbial taxa such as Faecalibacterium, Bacteroides and Romboutsia, which appear to be depleted in cancerogenic mucosa as well as in adenomatous polyps, thus representing novel microbial biomarkers associated with early tumor formation. Furthermore, an absolute quantification of Fusubacterium nucleatum in polyps further compounded the important role of this microorganism as a valuable putative microbial biomarker for early diagnosis of colorectal cancer.


Asunto(s)
Pólipos Adenomatosos/diagnóstico , Bacterias/clasificación , Biomarcadores/análisis , Neoplasias Colorrectales/diagnóstico , Pólipos Intestinales/diagnóstico , Microbiota/genética , Membrana Mucosa/metabolismo , Pólipos Adenomatosos/genética , Pólipos Adenomatosos/microbiología , Anciano , Bacterias/genética , Bacterias/aislamiento & purificación , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/microbiología , Femenino , Microbioma Gastrointestinal , Humanos , Pólipos Intestinales/genética , Pólipos Intestinales/microbiología , Masculino , Metagenómica , Membrana Mucosa/microbiología , Proyectos Piloto , Pronóstico , ARN Ribosómico 16S/genética
11.
Microbiome ; 6(1): 145, 2018 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-30126456

RESUMEN

BACKGROUND: Despite the relevance of viral populations, our knowledge of (bacterio) phage populations, i.e., the phageome, suffers from the absence of a "gold standard" protocol for viral DNA extraction with associated in silico sequence processing analyses. To overcome this apparent hiatus, we present here a comprehensive performance evaluation of various protocols and propose an optimized pipeline that covers DNA extraction, sequencing, and bioinformatic analysis of phageome data. RESULTS: Five widely used protocols for viral DNA extraction from fecal samples were tested for their performance in removal of non-viral DNA. Moreover, we developed a novel bioinformatic platform, METAnnotatorX, for metagenomic dataset analysis. This in silico tool facilitates a range of read- and assembly-based analyses, including taxonomic profiling using an iterative multi-database pipeline, classification of contigs at genus and species level, as well as functional characterizations of reads and assembled data. Performances of METAnnotatorX were assessed through investigation of seven mother-newborn pairs, leading to the identification of shared phage genotypes, of which two were genomically decoded and characterized. METAnnotatorX was furthermore employed to evaluate a protocol for the identification of contaminant non-viral DNA in sequenced datasets and was exploited to determine the amount of metagenomic data needed for robust evaluation of human adult-derived (fecal) phageomes. CONCLUSIONS: Results obtained in this study demonstrate that a comprehensive pipeline for analysis of phageomes will be pivotal for future explorations of the ecology of phages in the gut environment as well as for understanding their impact on the physiology and bacterial community kinetics as players of dysbiosis and homeostasis in the gut microbiota.


Asunto(s)
Bacteriófagos/clasificación , Biología Computacional/métodos , Heces/virología , Metagenómica/métodos , Bacteriófagos/genética , Bacteriófagos/aislamiento & purificación , Biología Computacional/normas , Femenino , Microbioma Gastrointestinal , Genotipo , Humanos , Recién Nacido , Masculino , Metagenómica/normas , Madres , Análisis de Secuencia de ADN/métodos , Programas Informáticos
12.
PLoS One ; 13(8): e0202670, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30161157

RESUMEN

Mesenteric ischemia/reperfusion is a clinical emergency with high morbidity and mortality due to the transient reduction of blood supply to the bowel. In recent years, the critical contribution of gut microbiome to human health and proper gastrointestinal functions has gradually emerged. In the current study, we investigated the protective effects of five days supplementation with Bifidobacterium bifidum PRL2010 in a murine model of gut ischemia/reperfusion. Our findings indicate that animals pretreated with B. bifidum PRL2010 showed lower neutrophil recruitment in the lungs, remarkably reduced bacterial translocation and decreased transcription levels of TNFalpha and IL-10 both in liver and kidneys, at the same time increasing those of IL-12 in kidneys. Inhibiting the adhesion of pathogenic bacteria and boosting host innate immunity responses are among the possible protective mechanisms enacted by the probiotic. These results demonstrate that short-period treatment with B. bifidum PRL2010 is a potential strategy to dampen remote organ injury due to mesenteric ischemia/reperfusion.


Asunto(s)
Bifidobacterium bifidum/fisiología , Intestinos/microbiología , Daño por Reperfusión/patología , Animales , Modelos Animales de Enfermedad , Heces/microbiología , Femenino , Inmunidad Innata , Interleucina-10/metabolismo , Intestinos/patología , Riñón/metabolismo , Hígado/metabolismo , Pulmón/inmunología , Pulmón/patología , Malondialdehído/metabolismo , Ratones , Neutrófilos/citología , Neutrófilos/inmunología , Probióticos/administración & dosificación , Daño por Reperfusión/inmunología , Daño por Reperfusión/prevención & control , Factor de Necrosis Tumoral alfa/metabolismo
13.
Cell Host Microbe ; 24(1): 133-145.e5, 2018 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-30001516

RESUMEN

The acquisition and development of the infant microbiome are key to establishing a healthy host-microbiome symbiosis. The maternal microbial reservoir is thought to play a crucial role in this process. However, the source and transmission routes of the infant pioneering microbes are poorly understood. To address this, we longitudinally sampled the microbiome of 25 mother-infant pairs across multiple body sites from birth up to 4 months postpartum. Strain-level metagenomic profiling showed a rapid influx of microbes at birth followed by strong selection during the first few days of life. Maternal skin and vaginal strains colonize only transiently, and the infant continues to acquire microbes from distinct maternal sources after birth. Maternal gut strains proved more persistent in the infant gut and ecologically better adapted than those acquired from other sources. Together, these data describe the mother-to-infant microbiome transmission routes that are integral in the development of the infant microbiome.


Asunto(s)
ADN Bacteriano/genética , Microbioma Gastrointestinal/genética , Tracto Gastrointestinal/microbiología , Relaciones Madre-Hijo , Adulto , Heces/microbiología , Femenino , Humanos , Lactante , Estudios Longitudinales , Metagenómica , Persona de Mediana Edad , Boca/microbiología , Piel/microbiología , Factores de Tiempo , Vagina/microbiología
14.
Appl Environ Microbiol ; 84(14)2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29728382

RESUMEN

The genus Lactobacillus is a widespread taxon, members of which are highly relevant to functional and fermented foods, while they are also commonly present in host-associated gut and vaginal microbiota. Substantial efforts have been undertaken to disclose the genetic repertoire of all members of the genus Lactobacillus, and yet their species-level profiling in complex matrices is still undeveloped due to the poor phylotype resolution of profiling approaches based on the 16S rRNA gene. To overcome this limitation, an internal transcribed spacer (ITS)-based profiling method was developed to accurately profile lactobacilli at the species level. This approach encompasses a genus-specific primer pair combined with a database of ITS sequences retrieved from all available Lactobacillus genomes and a script for the QIIME software suite that performs all required steps to reconstruct a species-level profile. This methodology was applied to several environments, i.e., human gut and vagina and the ceca of free-range chickens, as well as whey and fresh cheese. Interestingly, the data collected confirmed a relevant role of lactobacilli present in functional and fermented foods in defining the population harbored by the human gut, while, unsurprisingly perhaps, the ceca of free-range chickens were observed to be dominated by lactobacilli characterized in birds living in natural environments. Moreover, vaginal swabs confirmed the existence of previously hypothesized community state types, while analysis of whey and fresh cheese revealed a dominant presence of single Lactobacillus species used as starters for cheese production. Furthermore, application of this ITS profiling method to a mock Lactobacillus community allowed a minimal resolution level of <0.006 ng/µl.IMPORTANCE The genus Lactobacillus is a large and ubiquitous taxon of high scientific and commercial relevance. Despite the fact that the genetic repertoire of Lactobacillus species has been extensively characterized, the ecology of this genus has been explored by metataxonomic techniques that are accurate down to the genus or phylogenetic group level only. Thus, the distribution of lactobacilli in environmental or processed food samples is relatively unexplored. The profiling protocol described here relies on the use of the internal transcribed spacer to perform an accurate classification in a target population of lactobacilli with a <0.006-ng/µl sensitivity. This approach was used to analyze five sample types collected from both human and animal host-associated microbiota, as well as from the cheese production chain. The availability of a tool for species-level profiling of lactobacilli may be highly useful for both academic research and a wide range of industrial applications.


Asunto(s)
Lactobacillus/genética , Lactobacillus/metabolismo , Metagenómica , Animales , Queso/microbiología , Pollos , ADN Bacteriano/genética , Femenino , Microbioma Gastrointestinal , Perfilación de la Expresión Génica , Interacciones Microbiota-Huesped , Humanos , Lactobacillus/clasificación , ARN Ribosómico 16S/genética , Vagina/microbiología
15.
Gut ; 67(12): 2097-2106, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29705728

RESUMEN

OBJECTIVES: The involvement of the gut microbiota in the pathogenesis of calcium nephrolithiasis has been hypothesised since the discovery of the oxalate-degrading activity of Oxalobacter formigenes, but never comprehensively studied with metagenomics. The aim of this case-control study was to compare the faecal microbiota composition and functionality between recurrent idiopathic calcium stone formers (SFs) and controls. DESIGN: Faecal samples were collected from 52 SFs and 48 controls (mean age 48±11). The microbiota composition was analysed through 16S rRNA microbial profiling approach. Ten samples (five SFs, five controls) were also analysed with deep shotgun metagenomics sequencing, with focus on oxalate-degrading microbial metabolic pathways. Dietary habits, assessed through a food-frequency questionnaire, and 24-hour urinary excretion of prolithogenic and antilithogenic factors, including calcium and oxalate, were compared between SFs and controls, and considered as covariates in the comparison of microbiota profiles. RESULTS: SFs exhibited lower faecal microbial diversity than controls (Chao1 index 1460±363vs 1658±297, fully adjusted p=0.02 with stepwise backward regression analysis). At multivariate analyses, three taxa (Faecalibacterium, Enterobacter, Dorea) were significantly less represented in faecal samples of SFs. The Oxalobacter abundance was not different between groups. Faecal samples from SFs exhibited a significantly lower bacterial representation of genes involved in oxalate degradation, with inverse correlation with 24-hour oxalate excretion (r=-0.87, p=0.002). The oxalate-degrading genes were represented in several bacterial species, whose cumulative abundance was inversely correlated with oxaluria (r=-0.85, p=0.02). CONCLUSIONS: Idiopathic calcium SFs exhibited altered gut microbiota composition and functionality that could contribute to nephrolithiasis physiopathology.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Nefrolitiasis/microbiología , Adulto , Anciano , Bacterias/clasificación , Bacterias/aislamiento & purificación , Técnicas de Tipificación Bacteriana , Biodiversidad , Oxalato de Calcio/análisis , Estudios de Casos y Controles , ADN Bacteriano/análisis , Ingestión de Energía/fisiología , Heces/microbiología , Femenino , Humanos , Masculino , Metagenómica/métodos , Persona de Mediana Edad , Nefrolitiasis/metabolismo , Oxalatos/metabolismo , ARN Ribosómico 16S/análisis , Recurrencia , Adulto Joven
16.
Syst Appl Microbiol ; 41(3): 173-183, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29395537

RESUMEN

Six Bifidobacterium strains, i.e., Goo31D, Ham19E, Rab10A, Tam1G, Uis4E and Uis1B, were isolated from domestic goose (Anser domesticus), European hamster (Cricetus cricetus), European rabbit (Oryctolagus cuniculus), emperor tamarin (Saguinus imperator) and pygmy marmoset (Callithrix pygmaea). Cells are Gram-positive, non-motile, non-sporulating, facultative anaerobic and fructose 6-phosphate phosphoketolase-positive. Phylogenetic analyses based on 16S rRNA, ITS-, multilocus- sequences and the core genome revealed that bifidobacterial strains Goo31D, Ham19E, Rab10A, Tam1G, Uis4E and Uis1B exhibit close phylogenetic relatedness with Bifidobacterium choerinum LMG 10510, Bifidobacterium hapali DSM 100202, Bifidobacterium saguini DSM 23967 and Bifidobacterium stellenboschense DSM 23968. Genotyping based on the genome sequence of the isolated strains combined with phenotypic analyses, clearly show that these strains are distinct from each of the type strains of the so far recognized Bifidobacterium species. Thus, Bifidobacterium anseris sp. nov. (Goo31D=LMG 30189T=CCUG 70960T), Bifidobacterium criceti sp. nov. (Ham19E=LMG 30188T=CCUG 70962T), Bifidobacterium imperatoris sp. nov. (Tam1G=LMG 30297T=CCUG 70961T), Bifidobacterium italicum sp. nov. (Rab10A=LMG 30187T=CCUG 70963T), Bifidobacterium margollesii sp. nov. (Uis1B=LMG 30296T=CCUG 70959T) and Bifidobacterium parmae sp. nov. (Uis4E=LMG 30295T=CCUG 70964T) are proposed as novel Bifidobacterium species.


Asunto(s)
Bifidobacterium/clasificación , Heces/microbiología , Filogenia , Animales , Técnicas de Tipificación Bacteriana , Secuencia de Bases , Bifidobacterium/genética , Callithrix/microbiología , Cricetinae/microbiología , ADN Bacteriano/genética , Ácidos Grasos/análisis , Gansos/microbiología , Secuenciación de Nucleótidos de Alto Rendimiento , Italia , ARN Ribosómico 16S/genética , Conejos/microbiología , Saguinus/microbiología , Análisis de Secuencia de ADN
17.
Appl Environ Microbiol ; 84(4)2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29222102

RESUMEN

For decades, bacterial taxonomy has been based on in vitro molecular biology techniques and comparison of molecular marker sequences to measure the degree of genetic similarity and deduce phylogenetic relatedness of novel bacterial species to reference microbial taxa. Due to the advent of the genomic era, access to complete bacterial genome contents has become easier, thereby presenting the opportunity to precisely investigate the overall genetic diversity of microorganisms. Here, we describe a high-accuracy phylogenomic approach to assess the taxonomy of members of the genus Bifidobacterium and identify apparent misclassifications in current bifidobacterial taxonomy. The developed method was validated by the classification of seven novel taxa belonging to the genus Bifidobacterium by employing their overall genetic content. The results of this study demonstrate the potential of this whole-genome approach to become the gold standard for phylogenomics-based taxonomic classification of bacteria.IMPORTANCE Nowadays, next-generation sequencing has given access to genome sequences of the currently known bacterial taxa. The public databases constructed by means of these new technologies allowed comparison of genome sequences between microorganisms, providing information to perform genomic, phylogenomic, and evolutionary analyses. In order to avoid misclassifications in the taxonomy of novel bacterial isolates, new (bifido)bacterial taxons should be validated with a phylogenomic assessment like the approach presented here.


Asunto(s)
Bifidobacterium/clasificación , Bifidobacterium/genética , Genoma Bacteriano , Filogenia , Variación Genética , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , ARN Ribosómico 16S/genética
18.
Environ Microbiol ; 19(11): 4771-4783, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28967204

RESUMEN

Different factors may modulate the gut microbiota of animals. In any particular environment, diet, genetic factors and human influences can shape the bacterial communities residing in the gastrointestinal tract. Metagenomic approaches have significantly expanded our knowledge on microbiota dynamics inside hosts, yet cultivation and isolation of bacterial members of these complex ecosystems may still be necessary to fully understand interactions between bacterial communities and their host. A dual approach, involving culture-independent and -dependent techniques, was used here to decipher the microbiota communities that inhabit the gastro intestinal tract of free-range, broiler and feral chickens. In silico analysis revealed the presence of a core microbiota that is typical of those animals that live in different geographical areas and that have limited contact with humans. Anthropic influences guide the metabolic potential and the presence of antibiotic resistance genes of these different bacterial communities. Culturomics attempts, based on different cultivation conditions, were applied to reconstruct in vitro the microbiota of feral chickens. A unique strain collection representing members of the four major phyla of the poultry microbiota was assembled, including bacterial strains that are not typically retrieved from the chicken gut.


Asunto(s)
Bacterias/clasificación , Bacterias/aislamiento & purificación , Ciego/microbiología , Pollos/microbiología , Microbioma Gastrointestinal/genética , Animales , Bacterias/genética , Dieta , Geografía , Humanos , Metagenómica
19.
Sci Rep ; 7(1): 11102, 2017 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-28894183

RESUMEN

Reduced biodiversity and increased representation of opportunistic pathogens are typical features of gut microbiota composition in aging. Few studies have investigated their correlation with polypharmacy, multimorbidity and frailty. To assess it, we analyzed the fecal microbiota from 76 inpatients, aged 83 ± 8. Microbiome biodiversity (Chao1 index) and relative abundance of individual bacterial taxa were determined by next-generation 16S rRNA microbial profiling. Their correlation with number of drugs, and indexes of multimorbidity and frailty were verified using multivariate linear regression models. The impact of gut microbiota biodiversity on mortality, rehospitalizations and incident sepsis was also assessed after a 2-year follow-up, using Cox regression analysis. We found a significant negative correlation between the number of drugs and Chao1 Index at multivariate analysis. The number of drugs was associated with the average relative abundance of 15 taxa. The drug classes exhibiting the strongest association with single taxa abundance were proton pump inhibitors, antidepressants and antipsychotics. Conversely, frailty and multimorbidity were not significantly associated with gut microbiota biodiversity. Very low Chao1 index was also a significant predictor of mortality, but not of rehospitalizations and sepsis, at follow-up. In aging, polypharmacy may thus represent a determinant of gut microbiota composition, with detrimental clinical consequences.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Hospitalización , Polifarmacia , Anciano , Anciano de 80 o más Años , Biodiversidad , Variación Biológica Poblacional , Comorbilidad , Femenino , Humanos , Masculino , Metagenoma , Metagenómica , Evaluación del Resultado de la Atención al Paciente , Fenotipo , ARN Ribosómico 16S , Análisis de Supervivencia , Evaluación de Síntomas
20.
Front Microbiol ; 8: 1749, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28955319

RESUMEN

The composition of the gut microbiota of mammals is greatly influenced by diet. Therefore, evaluation of different food ingredients that may promote changes in the gut microbiota composition is an attractive approach to treat microbiota disturbances. In this study, three dietary fibers, such as inulin (I, 10%), resistant starch (RS, 10%), and citrus pectin (3%), were employed as supplements to normal chow diet of adult male rats for 2 weeks. Fecal microbiota composition and corresponding metabolite profiles were assessed before and after prebiotics supplementation. A general increase in the Bacteroidetes phylum was detected with a concurrent reduction in Firmicutes, in particular for I and RS experiments, while additional changes in the microbiota composition were evident at lower taxonomic levels for all the three substrates. Such modifications in the microbiota composition were correlated with changes in metabolic profiles of animals, in particular changes in acetate and succinate levels. This study represents a first attempt to modulate selectively the abundance and/or metabolic activity of various members of the gut microbiota by means of dietary fiber.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...