Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 6044, 2022 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-36229545

RESUMEN

Despite extensive efforts to address it, the vastness of uncharacterized 'dark matter' microbial genetic diversity can impact short-read sequencing based metagenomic studies. Population-specific biases in genomic reference databases can further compound this problem. Leveraging advances in hybrid assembly (using short and long reads) and Hi-C technologies in a cross-sectional survey, we deeply characterized 109 gut microbiomes from three ethnicities in Singapore to comprehensively reconstruct 4497 medium and high-quality metagenome assembled genomes, 1708 of which were missing in short-read only analysis and with >28× N50 improvement. Species-level clustering identified 70 (>10% of total) novel gut species out of 685, improved reference genomes for 363 species (53% of total), and discovered 3413 strains unique to these populations. Among the top 10 most abundant gut bacteria in our study, one of the species and >80% of strains were unrepresented in existing databases. Annotation of biosynthetic gene clusters (BGCs) uncovered more than 27,000 BGCs with a large fraction (36-88%) unrepresented in current databases, and with several unique clusters predicted to produce bacteriocins that could significantly alter microbiome community structure. These results reveal significant uncharacterized gut microbial diversity in Southeast Asian populations and highlight the utility of hybrid metagenomic references for bioprospecting and disease-focused studies.


Asunto(s)
Bacteriocinas , Microbiota , Pueblo Asiatico/genética , Bacteriocinas/genética , Estudios Transversales , Genoma Humano , Humanos , Metagenoma/genética , Metagenómica/métodos , Microbiota/genética
2.
J Allergy Clin Immunol ; 147(4): 1329-1340, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33039480

RESUMEN

BACKGROUND: Atopic dermatitis (AD) is a common skin disease affecting up to 20% of the global population, with significant clinical heterogeneity and limited information about molecular subtypes and actionable biomarkers. Although alterations in the skin microbiome have been described in subjects with AD during progression to flare state, the prognostic value of baseline microbiome configurations has not been explored. OBJECTIVE: Our aim was to identify microbial signatures on AD skin that are predictive of disease fate. METHODS: Nonlesional skin of patients with AD and healthy control subjects were sampled at 2 time points separated by at least 4 weeks. Using whole metagenome analysis of skin microbiomes of patients with AD and control subjects (n = 49 and 189 samples), we identified distinct microbiome configurations (dermotypes A and B). Blood was collected for immunophenotyping, and skin surface samples were analyzed for correlations with natural moisturizing factors and antimicrobial peptides. RESULTS: Dermotypes were robust and validated across 2 additional cohorts (63 individuals), with strong enrichment of subjects with AD in dermotype B. Dermotype B was characterized by reduced microbial richness, depletion of Cutibacterium acnes, Dermacoccus and Methylobacterium species, individual-specific outlier abundance of Staphylococcus species (eg, S epidermidis, S capitis, S aureus), and enrichment in metabolic pathways (eg, branched chain amino acids and arginine biosynthesis) and virulence genes (eg, ß-toxin, δ-toxin) that defined a pathogenic ecology. Skin surface and circulating host biomarkers exhibited a distinct microbial-associated signature that was further reflected in more severe itching, frequent flares, and increased disease severity in patients harboring the dermotype B microbiome. CONCLUSION: We report distinct clusters of microbial profiles that delineate the role of microbiome configurations in AD heterogeneity, highlight a mechanism for ongoing inflammation, and provide prognostic utility toward microbiome-based disease stratification.


Asunto(s)
Dermatitis Atópica/microbiología , Microbiota , Piel/microbiología , Adolescente , Adulto , Bacterias/genética , Bacterias/patogenicidad , Biomarcadores/sangre , Citocinas/sangre , Dermatitis Atópica/sangre , Dermatitis Atópica/inmunología , Dermatitis Atópica/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Índice de Severidad de la Enfermedad , Piel/química , Piel/metabolismo , Pruebas Cutáneas , Virulencia/genética , Agua/metabolismo , Adulto Joven
3.
Nat Ecol Evol ; 4(9): 1256-1267, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32632261

RESUMEN

Loss of diversity in the gut microbiome can persist for extended periods after antibiotic treatment, impacting microbiome function, antimicrobial resistance and probably host health. Despite widespread antibiotic use, our understanding of the species and metabolic functions contributing to gut microbiome recovery is limited. Using data from 4 discovery cohorts in 3 continents comprising >500 microbiome profiles from 117 individuals, we identified 21 bacterial species exhibiting robust association with ecological recovery post antibiotic therapy. Functional and growth-rate analysis showed that recovery is supported by enrichment in specific carbohydrate-degradation and energy-production pathways. Association rule mining on 782 microbiome profiles from the MEDUSA database enabled reconstruction of the gut microbial 'food web', identifying many recovery-associated bacteria as keystone species, with the ability to use host- and diet-derived energy sources, and support repopulation of other gut species. Experiments in a mouse model recapitulated the ability of recovery-associated bacteria (Bacteroides thetaiotaomicron and Bifidobacterium adolescentis) to promote recovery with synergistic effects, providing a boost of two orders of magnitude to microbial abundance in early time points and faster maturation of microbial diversity. The identification of specific species and metabolic functions promoting recovery opens up opportunities for rationally determining pre- and probiotic formulations offering protection from long-term consequences of frequent antibiotic usage.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Animales , Antibacterianos , Bacterias/genética , Humanos , Metagenoma , Ratones
4.
Nat Med ; 26(6): 941-951, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32514171

RESUMEN

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.


Asunto(s)
Infección Hospitalaria/microbiología , Farmacorresistencia Bacteriana/genética , Equipos y Suministros de Hospitales/microbiología , Control de Infecciones , Microbiota/genética , Lechos/microbiología , Biopelículas , Infección Hospitalaria/tratamiento farmacológico , Infección Hospitalaria/transmisión , Desinfección , Farmacorresistencia Bacteriana Múltiple/genética , Contaminación de Equipos , Mapeo Geográfico , Humanos , Metagenómica , Infecciones Oportunistas/tratamiento farmacológico , Infecciones Oportunistas/microbiología , Infecciones Oportunistas/transmisión , Habitaciones de Pacientes , Singapur , Análisis Espacio-Temporal , Centros de Atención Terciaria
5.
Nat Biotechnol ; 37(8): 937-944, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31359005

RESUMEN

Characterization of microbiomes has been enabled by high-throughput metagenomic sequencing. However, existing methods are not designed to combine reads from short- and long-read technologies. We present a hybrid metagenomic assembler named OPERA-MS that integrates assembly-based metagenome clustering with repeat-aware, exact scaffolding to accurately assemble complex communities. Evaluation using defined in vitro and virtual gut microbiomes revealed that OPERA-MS assembles metagenomes with greater base pair accuracy than long-read (>5×; Canu), higher contiguity than short-read (~10× NGA50; MEGAHIT, IDBA-UD, metaSPAdes) and fewer assembly errors than non-metagenomic hybrid assemblers (2×; hybridSPAdes). OPERA-MS provides strain-resolved assembly in the presence of multiple genomes of the same species, high-quality reference genomes for rare species (<1%) with ~9× long-read coverage and near-complete genomes with higher coverage. We used OPERA-MS to assemble 28 gut metagenomes of antibiotic-treated patients, and showed that the inclusion of long nanopore reads produces more contiguous assemblies (200× improvement over short-read assemblies), including more than 80 closed plasmid or phage sequences and a new 263 kbp jumbo phage. High-quality hybrid assemblies enable an exquisitely detailed view of the gut resistome in human patients.


Asunto(s)
Bacterias/efectos de los fármacos , Bacterias/genética , Metagenómica/métodos , Microbiota/efectos de los fármacos , Análisis de Secuencia de ADN/métodos , Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Heces/microbiología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Metagenoma , Nanoporos , Programas Informáticos
6.
Microbiome ; 6(1): 213, 2018 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-30497517

RESUMEN

BACKGROUND: Even though human sweat is odorless, bacterial growth and decomposition of specific odor precursors in it is believed to give rise to body odor in humans. While mechanisms of odor generation have been widely studied in adults, little is known for teenagers and pre-pubescent children who have distinct sweat composition from immature apocrine and sebaceous glands, but are arguably more susceptible to the social and psychological impact of malodor. RESULTS: We integrated information from whole microbiome analysis of multiple skin sites (underarm, neck, and head) and multiple time points (1 h and 8 h after bath), analyzing 180 samples in total to perform the largest metagenome-wide association study to date on malodor. Significant positive correlations were observed between odor intensity and the relative abundance of Staphylococcus hominis, Staphylococcus epidermidis, and Cutibacterium avidum, as well as negative correlation with Acinetobacter schindleri and Cutibacterium species. Metabolic pathway analysis highlighted the association of isovaleric and acetic acid production (sour odor) from enriched S. epidermidis (teen underarm) and S. hominis (child neck) enzymes and sulfur production from Staphylococcus species (teen underarm) with odor intensity, in good agreement with observed odor characteristics in pre-pubescent children and teenagers. Experiments with cultures on human and artificial sweat confirmed the ability of S. hominis and S. epidermidis to independently produce malodor with distinct odor characteristics. CONCLUSIONS: These results showcase the power of skin metagenomics to study host-microbial co-metabolic interactions, identifying distinct pathways for odor generation from sweat in pre-pubescent children and teenagers and highlighting key enzymatic targets for intervention.


Asunto(s)
Bacterias/clasificación , Metagenómica/métodos , Odorantes/análisis , Piel/microbiología , Sudor/microbiología , Ácido Acético/análisis , Acinetobacter/clasificación , Acinetobacter/aislamiento & purificación , Adolescente , Axila/microbiología , Bacterias/aislamiento & purificación , Niño , Femenino , Cabeza/microbiología , Hemiterpenos , Humanos , Masculino , Cuello/microbiología , Ácidos Pentanoicos/análisis , Propionibacteriaceae/clasificación , Propionibacteriaceae/aislamiento & purificación , Pubertad , Análisis de Secuencia de ADN , Piel/química , Staphylococcus epidermidis/clasificación , Staphylococcus epidermidis/aislamiento & purificación , Staphylococcus hominis/clasificación , Staphylococcus hominis/aislamiento & purificación , Azufre/análisis
7.
Hepatology ; 67(1): 282-295, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28646502

RESUMEN

The gut microbiota possesses diverse metabolic activities, but its contribution toward heterogeneous toxicological responses is poorly understood. In this study, we investigated the role of the liver-gut microbiota axis in underpinning the hepatotoxicity of tacrine. We employed an integrated strategy combining pharmacokinetics, toxicology, metabonomics, genomics, and metagenomics to elucidate and validate the mechanism of tacrine-induced hepatotoxicity in Lister hooded rats. Pharmacokinetic studies in rats demonstrated 3.3-fold higher systemic exposure to tacrine in strong responders that experienced transaminitis, revealing enhanced enterohepatic recycling of deglucuronidated tacrine in this subgroup, not attributable to variation in hepatic disposition gene expression. Metabonomic studies implicated variations in gut microbial activities that mapped onto tacrine-induced transaminitis. Metagenomics delineated greater deglucuronidation capabilities in strong responders, based on differential gut microbial composition (e.g., Lactobacillus, Bacteroides, and Enterobacteriaceae) and approximately 9% higher ß-glucuronidase gene abundance compared with nonresponders. In the validation study, coadministration with oral ß-glucuronidase derived from Escherichia coli and pretreatment with vancomycin and imipenem significantly modulated the susceptibility to tacrine-induced transaminitis in vivo. CONCLUSION: This study establishes pertinent gut microbial influences in modifying the hepatotoxicity of tacrine, providing insights for personalized medicine initiatives. (Hepatology 2018;67:282-295).


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Microbioma Gastrointestinal/efectos de los fármacos , Tacrina/toxicidad , Animales , Biopsia con Aguja , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inmunohistoquímica , Pruebas de Función Hepática , Masculino , Distribución Aleatoria , Ratas , Ratas Endogámicas , Valores de Referencia , Índice de Severidad de la Enfermedad , Tacrina/farmacocinética , Tacrina/farmacología
8.
Gigascience ; 5(1): 34, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27485345

RESUMEN

BACKGROUND: Nanopore sequencing provides a rapid, cheap and portable real-time sequencing platform with the potential to revolutionize genomics. However, several applications are limited by relatively high single-read error rates (>10 %), including RNA-seq, haplotype sequencing and 16S sequencing. RESULTS: We developed the Intramolecular-ligated Nanopore Consensus Sequencing (INC-Seq) as a strategy for obtaining long and accurate nanopore reads, starting with low input DNA. Applying INC-Seq for 16S rRNA-based bacterial profiling generated full-length amplicon sequences with a median accuracy >97 %. CONCLUSIONS: INC-Seq reads enabled accurate species-level classification, identification of species at 0.1 % abundance and robust quantification of relative abundances, providing a cheap and effective approach for pathogen detection and microbiome profiling on the MinION system.


Asunto(s)
Bacterias/clasificación , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN/métodos , Algoritmos , Bacterias/genética , Código de Barras del ADN Taxonómico , ADN Bacteriano/genética , ADN Ribosómico/genética , Genómica , Humanos , Nanoporos
9.
Nat Microbiol ; 1(9): 16106, 2016 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-27562258

RESUMEN

Whole metagenome analysis has the potential to reveal functional triggers of skin diseases, but issues of cost, robustness and sampling efficacy have limited its application. Here, we have established an alternative, clinically practical and robust metagenomic analysis protocol and applied it to 80 skin microbiome samples epidemiologically stratified for atopic dermatitis (AD). We have identified distinct non-flare, baseline skin microbiome signatures enriched for Streptococcus and Gemella but depleted for Dermacoccus in AD-prone versus normal healthy skin. Bacterial challenge assays using keratinocytes and monocyte-derived dendritic cells established distinct IL-1-mediated, innate and Th1-mediated adaptive immune responses with Staphylococcus aureus and Staphylococcus epidermidis. Bacterial differences were complemented by perturbations in the eukaryotic community and functional shifts in the microbiome-wide gene repertoire, which could exacerbate a dry and alkaline phenotype primed for pathogen growth and inflammation in AD-susceptible skin. These findings provide insights into how the skin microbial community, skin surface microenvironment and immune system cross-modulate each other, escalating the destructive feedback cycle between them that leads to AD flare.


Asunto(s)
Dermatitis Atópica/microbiología , Metagenoma , Microbiota/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/inmunología , Staphylococcus epidermidis/inmunología , Inmunidad Adaptativa , Adulto , Animales , Células Dendríticas/patología , Dermatitis Atópica/inmunología , Susceptibilidad a Enfermedades , Femenino , Humanos , Interleucina-1/inmunología , Masculino , Metagenómica , Ratones Endogámicos C57BL , Piel/inmunología , Infecciones Estafilocócicas/inmunología , Adulto Joven
10.
EBioMedicine ; 8: 195-202, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27428430

RESUMEN

Cholangiocarcinoma (CCA) is the primary cancer of the bile duct system. The role of bile duct tissue microbiomes in CCA tumorigenesis is unestablished. To address this, sixty primary CCA tumors and matched normals, from both liver fluke (Opisthorchis viverrini) associated (OVa, n=28) and non-O. viverrini associated (non-OVa, n=32) cancers, were profiled using high-throughput 16S rRNA sequencing. A distinct, tissue-specific microbiome dominated by the bacterial families Dietziaceae, Pseudomonadaceae and Oxalobacteraceae was observed in bile duct tissues. Systemic perturbation of the microbiome was noted in tumor and paired normal samples (vs non-cancer normals) for several bacterial families with a significant increase in Stenotrophomonas species distinguishing tumors vs paired normals. Comparison of parasite associated (OVa) vs non-associated (non-OVa) groups identified enrichment for specific enteric bacteria (Bifidobacteriaceae, Enterobacteriaceae and Enterococcaceae). One of the enriched families, Bifidobacteriaceae, was found to be dominant in the O. viverrini microbiome, providing a mechanistic link to the parasite. Functional analysis and comparison of CCA microbiomes revealed higher potential for producing bile acids and ammonia in OVa tissues, linking the altered microbiota to carcinogenesis. These results define how the unique microbial communities resident in the bile duct, parasitic infections and the tissue microenvironment can influence each other, and contribute to cancer.


Asunto(s)
Neoplasias de los Conductos Biliares/etiología , Colangiocarcinoma/etiología , Microbioma Gastrointestinal , Microbiota , Opistorquiasis/complicaciones , Opistorquiasis/parasitología , Opisthorchis , Adulto , Anciano , Animales , Biodiversidad , Transformación Celular Neoplásica , Femenino , Humanos , Masculino , Metagenoma , Metagenómica/métodos , Persona de Mediana Edad , Especificidad de Órganos , ARN Ribosómico 16S
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...