Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Comput Biol ; 17(9): e1009343, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34495960

RESUMO

CONCLUSION: BEEM-Static provides new opportunities for mining ecologically interpretable interactions and systems insights from the growing corpus of microbiome data.


Assuntos
Ecossistema , Microbioma Gastrointestinal , Biomassa , Estudos Transversais , Conjuntos de Dados como Assunto , Humanos
2.
Nat Med ; 26(6): 941-951, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32514171

RESUMO

Although disinfection is key to infection control, the colonization patterns and resistomes of hospital-environment microbes remain underexplored. We report the first extensive genomic characterization of microbiomes, pathogens and antibiotic resistance cassettes in a tertiary-care hospital, from repeated sampling (up to 1.5 years apart) of 179 sites associated with 45 beds. Deep shotgun metagenomics unveiled distinct ecological niches of microbes and antibiotic resistance genes characterized by biofilm-forming and human-microbiome-influenced environments with corresponding patterns of spatiotemporal divergence. Quasi-metagenomics with nanopore sequencing provided thousands of high-contiguity genomes, phage and plasmid sequences (>60% novel), enabling characterization of resistome and mobilome diversity and dynamic architectures in hospital environments. Phylogenetics identified multidrug-resistant strains as being widely distributed and stably colonizing across sites. Comparisons with clinical isolates indicated that such microbes can persist in hospitals for extended periods (>8 years), to opportunistically infect patients. These findings highlight the importance of characterizing antibiotic resistance reservoirs in hospitals and establish the feasibility of systematic surveys to target resources for preventing infections.


Assuntos
Infecção Hospitalar/microbiologia , Farmacorresistência Bacteriana/genética , Equipamentos e Provisões Hospitalares/microbiologia , Controle de Infecções , Microbiota/genética , Leitos/microbiologia , Biofilmes , Infecção Hospitalar/tratamento farmacológico , Infecção Hospitalar/transmissão , Desinfecção , Farmacorresistência Bacteriana Múltipla/genética , Contaminação de Equipamentos , Mapeamento Geográfico , Humanos , Metagenômica , Infecções Oportunistas/tratamento farmacológico , Infecções Oportunistas/microbiologia , Infecções Oportunistas/transmissão , Quartos de Pacientes , Singapura , Análise Espaço-Temporal , Centros de Atenção Terciária
3.
Microbiome ; 7(1): 118, 2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31439018

RESUMO

BACKGROUND: The dynamics of microbial communities is driven by a range of interactions from symbiosis to predator-prey relationships, the majority of which are poorly understood. With the increasing availability of high-throughput microbiome taxonomic profiling data, it is now conceivable to directly learn the ecological models that explicitly define microbial interactions and explain community dynamics. The applicability of these approaches is severely limited by the lack of accurate absolute cell density measurements (biomass). METHODS: We present a new computational approach that resolves this key limitation in the inference of generalized Lotka-Volterra models (gLVMs) by coupling biomass estimation and model inference with an expectation-maximization algorithm (BEEM). RESULTS: BEEM outperforms the state-of-the-art methods for inferring gLVMs, while simultaneously eliminating the need for additional experimental biomass data as input. BEEM's application to previously inaccessible public datasets (due to the lack of biomass data) allowed us to construct ecological models of microbial communities in the human gut on a per-individual basis, revealing personalized dynamics and keystone species. CONCLUSIONS: BEEM addresses a key bottleneck in "systems analysis" of microbiomes by enabling accurate inference of ecological models from high throughput sequencing data without the need for experimental biomass measurements.


Assuntos
Algoritmos , Microbioma Gastrointestinal/fisiologia , Interações Microbianas , Modelos Biológicos , Conjuntos de Dados como Assunto , Microbioma Gastrointestinal/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA