Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 21(3): 343-353, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32066951

RESUMEN

Gastrointestinal microbiota and immune cells interact closely and display regional specificity; however, little is known about how these communities differ with location. Here, we simultaneously assess microbiota and single immune cells across the healthy, adult human colon, with paired characterization of immune cells in the mesenteric lymph nodes, to delineate colonic immune niches at steady state. We describe distinct helper T cell activation and migration profiles along the colon and characterize the transcriptional adaptation trajectory of regulatory T cells between lymphoid tissue and colon. Finally, we show increasing B cell accumulation, clonal expansion and mutational frequency from the cecum to the sigmoid colon and link this to the increasing number of reactive bacterial species.


Asunto(s)
Colon/inmunología , Colon/microbiología , Microbioma Gastrointestinal/inmunología , Adulto , Linfocitos B/inmunología , Colon/citología , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Especificidad de Órganos , RNA-Seq , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Transcriptoma
2.
Nat Immunol ; 14(9): 901-7, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23872679

RESUMEN

Type I interferons are important in regulating immune responses to pathogens and tumors. All interferons are considered to signal via the heterodimeric IFNAR1-IFNAR2 complex, yet some subtypes such as interferon-ß (IFN-ß) can exhibit distinct functional properties, although the molecular basis of this is unclear. Here we demonstrate IFN-ß can uniquely and specifically ligate to IFNAR1 in an IFNAR2-independent manner, and we provide the structural basis of the IFNAR1-IFN-ß interaction. The IFNAR1-IFN-ß complex transduced signals that modulated expression of a distinct set of genes independently of Jak-STAT pathways. Lipopolysaccharide-induced sepsis was ameliorated in Ifnar1(-/-) mice but not Ifnar2(-/-) mice, suggesting that IFNAR1-IFN-ß signaling is pathologically relevant. Thus, we provide a molecular basis for understanding specific functions of IFN-ß.


Asunto(s)
Interferón beta/química , Interferón beta/metabolismo , Receptor de Interferón alfa y beta/química , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Femenino , Lipopolisacáridos/efectos adversos , Ratones , Ratones Noqueados , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Unión Proteica , Conformación Proteica , Estabilidad Proteica , Receptor de Interferón alfa y beta/genética , Choque Séptico/inducido químicamente , Choque Séptico/genética , Choque Séptico/metabolismo , Choque Séptico/mortalidad
3.
Nature ; 568(7753): 499-504, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30745586

RESUMEN

The composition of the human gut microbiota is linked to health and disease, but knowledge of individual microbial species is needed to decipher their biological roles. Despite extensive culturing and sequencing efforts, the complete bacterial repertoire of the human gut microbiota remains undefined. Here we identify 1,952 uncultured candidate bacterial species by reconstructing 92,143 metagenome-assembled genomes from 11,850 human gut microbiomes. These uncultured genomes substantially expand the known species repertoire of the collective human gut microbiota, with a 281% increase in phylogenetic diversity. Although the newly identified species are less prevalent in well-studied populations compared to reference isolate genomes, they improve classification of understudied African and South American samples by more than 200%. These candidate species encode hundreds of newly identified biosynthetic gene clusters and possess a distinctive functional capacity that might explain their elusive nature. Our work expands the known diversity of uncultured gut bacteria, which provides unprecedented resolution for taxonomic and functional characterization of the intestinal microbiota.


Asunto(s)
Bacterias/clasificación , Bacterias/genética , Microbioma Gastrointestinal/genética , Genoma Bacteriano/genética , Genómica , Metagenoma/genética , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Humanos , Familia de Multigenes , Filogenia , Especificidad de la Especie
4.
Nature ; 574(7776): 117-121, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31534227

RESUMEN

Immediately after birth, newborn babies experience rapid colonization by microorganisms from their mothers and the surrounding environment1. Diseases in childhood and later in life are potentially mediated by the perturbation of the colonization of the infant gut microbiota2. However, the effects of delivery via caesarean section on the earliest stages of the acquisition and development of the gut microbiota, during the neonatal period (≤1 month), remain controversial3,4. Here we report the disrupted transmission of maternal Bacteroides strains, and high-level colonization by opportunistic pathogens associated with the hospital environment (including Enterococcus, Enterobacter and Klebsiella species), in babies delivered by caesarean section. These effects were also seen, to a lesser extent, in vaginally delivered babies whose mothers underwent antibiotic prophylaxis and in babies who were not breastfed during the neonatal period. We applied longitudinal sampling and whole-genome shotgun metagenomic analysis to 1,679 gut microbiota samples (taken at several time points during the neonatal period, and in infancy) from 596 full-term babies born in UK hospitals; for a subset of these babies, we collected additional matched samples from mothers (175 mothers paired with 178 babies). This analysis demonstrates that the mode of delivery is a significant factor that affects the composition of the gut microbiota throughout the neonatal period, and into infancy. Matched large-scale culturing and whole-genome sequencing of over 800 bacterial strains from these babies identified virulence factors and clinically relevant antimicrobial resistance in opportunistic pathogens that may predispose individuals to opportunistic infections. Our findings highlight the critical role of the local environment in establishing the gut microbiota in very early life, and identify colonization with antimicrobial-resistance-containing opportunistic pathogens as a previously underappreciated risk factor in hospital births.


Asunto(s)
Cesárea/efectos adversos , Microbioma Gastrointestinal , Enfermedades del Recién Nacido/microbiología , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Infecciones Oportunistas/congénito , Infecciones Oportunistas/microbiología , Femenino , Humanos , Recién Nacido , Enfermedades del Recién Nacido/etiología , Infecciones Oportunistas/etiología , Embarazo
5.
Gut ; 73(5): 751-769, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38331563

RESUMEN

OBJECTIVE: Chronic obstructive pulmonary disease (COPD) is a major cause of global illness and death, most commonly caused by cigarette smoke. The mechanisms of pathogenesis remain poorly understood, limiting the development of effective therapies. The gastrointestinal microbiome has been implicated in chronic lung diseases via the gut-lung axis, but its role is unclear. DESIGN: Using an in vivo mouse model of cigarette smoke (CS)-induced COPD and faecal microbial transfer (FMT), we characterised the faecal microbiota using metagenomics, proteomics and metabolomics. Findings were correlated with airway and systemic inflammation, lung and gut histopathology and lung function. Complex carbohydrates were assessed in mice using a high resistant starch diet, and in 16 patients with COPD using a randomised, double-blind, placebo-controlled pilot study of inulin supplementation. RESULTS: FMT alleviated hallmark features of COPD (inflammation, alveolar destruction, impaired lung function), gastrointestinal pathology and systemic immune changes. Protective effects were additive to smoking cessation, and transfer of CS-associated microbiota after antibiotic-induced microbiome depletion was sufficient to increase lung inflammation while suppressing colonic immunity in the absence of CS exposure. Disease features correlated with the relative abundance of Muribaculaceae, Desulfovibrionaceae and Lachnospiraceae family members. Proteomics and metabolomics identified downregulation of glucose and starch metabolism in CS-associated microbiota, and supplementation of mice or human patients with complex carbohydrates improved disease outcomes. CONCLUSION: The gut microbiome contributes to COPD pathogenesis and can be targeted therapeutically.


Asunto(s)
Neumonía , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Ratones , Animales , Enfermedad Pulmonar Obstructiva Crónica/etiología , Pulmón/metabolismo , Pulmón/patología , Neumonía/etiología , Inflamación/metabolismo , Carbohidratos/farmacología
6.
Bioinformatics ; 38(20): 4814-4816, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36029242

RESUMEN

SUMMARY: Shotgun metagenomic sequencing provides the capacity to understand microbial community structure and function at unprecedented resolution; however, the current analytical methods are constrained by a focus on taxonomic classifications that may obfuscate functional relationships. Here, we present expam, a tree-based, taxonomy agnostic tool for the identification of biologically relevant clades from shotgun metagenomic sequencing. AVAILABILITY AND IMPLEMENTATION: expam is an open-source Python application released under the GNU General Public Licence v3.0. expam installation instructions, source code and tutorials can be found at https://github.com/seansolari/expam. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Metagenoma , Microbiota , Metagenómica/métodos , Microbiota/genética , Programas Informáticos
7.
Nature ; 533(7604): 543-546, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27144353

RESUMEN

Our intestinal microbiota harbours a diverse bacterial community required for our health, sustenance and wellbeing. Intestinal colonization begins at birth and climaxes with the acquisition of two dominant groups of strict anaerobic bacteria belonging to the Firmicutes and Bacteroidetes phyla. Culture-independent, genomic approaches have transformed our understanding of the role of the human microbiome in health and many diseases. However, owing to the prevailing perception that our indigenous bacteria are largely recalcitrant to culture, many of their functions and phenotypes remain unknown. Here we describe a novel workflow based on targeted phenotypic culturing linked to large-scale whole-genome sequencing, phylogenetic analysis and computational modelling that demonstrates that a substantial proportion of the intestinal bacteria are culturable. Applying this approach to healthy individuals, we isolated 137 bacterial species from characterized and candidate novel families, genera and species that were archived as pure cultures. Whole-genome and metagenomic sequencing, combined with computational and phenotypic analysis, suggests that at least 50-60% of the bacterial genera from the intestinal microbiota of a healthy individual produce resilient spores, specialized for host-to-host transmission. Our approach unlocks the human intestinal microbiota for phenotypic analysis and reveals how a marked proportion of oxygen-sensitive intestinal bacteria can be transmitted between individuals, affecting microbiota heritability.


Asunto(s)
Bacterias/clasificación , Bacterias/crecimiento & desarrollo , Técnicas de Tipificación Bacteriana , Microbioma Gastrointestinal/fisiología , Anaerobiosis , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/aislamiento & purificación , Técnicas de Cultivo de Célula , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Genoma Bacteriano/genética , Salud , Humanos , Metagenoma/genética , Metagenómica , Oxígeno/metabolismo , Oxígeno/farmacología , Fenotipo , Filogenia , Análisis de Secuencia de ADN , Especificidad de la Especie , Esporas Bacterianas/clasificación , Esporas Bacterianas/efectos de los fármacos , Esporas Bacterianas/crecimiento & desarrollo
8.
Nutr Res Rev ; 35(2): 207-220, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-34100344

RESUMEN

There is mounting evidence that microbiome composition is intimately and dynamically connected with host energy balance and metabolism. The gut microbiome is emerging as a novel target for counteracting the chronically positive energy balance in obesity, a disease of pandemic scale which contributes to >70 % of premature deaths. This scoping review explores the potential for therapeutic modulation of gut microbiota as a means of prevention and/or treatment of obesity and obesity-associated metabolic disorders. The evidence base for interventional approaches which have been shown to affect the composition and function of the intestinal microbiome is summarised, including dietary strategies, oral probiotic treatment, faecal microbiota transplantation and bariatric surgery. Evidence in this field is still largely derived from preclinical rodent models, but interventional studies in obese populations have demonstrated metabolic improvements effected by microbiome-modulating treatments such as faecal microbiota transplantation, as well as drawing attention to the unappreciated role of microbiome modulation in well-established anti-obesity interventions, such as dietary change or bariatric surgery. The complex relationship between microbiome composition and host metabolism will take time to unravel, but microbiome modulation is likely to provide a novel strategy in the limited armamentarium of effective treatments for obesity.


Asunto(s)
Microbioma Gastrointestinal , Probióticos , Humanos , Prebióticos , Obesidad/terapia , Obesidad/metabolismo , Trasplante de Microbiota Fecal , Probióticos/uso terapéutico
9.
Allergy ; 76(3): 714-734, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32762040

RESUMEN

The prevalence of chronic immune and metabolic disorders is increasing rapidly. In particular, inflammatory bowel diseases, obesity, diabetes, asthma and chronic obstructive pulmonary disease have become major healthcare and economic burdens worldwide. Recent advances in microbiome research have led to significant discoveries of associative links between alterations in the microbiome and health, as well as these chronic supposedly noncommunicable, immune/metabolic disorders. Importantly, the interplay between diet, microbiome and the mucous barrier in these diseases has gained significant attention. Diet modulates the mucous barrier via alterations in gut microbiota, resulting in either disease onset/exacerbation due to a "poor" diet or protection against disease with a "healthy" diet. In addition, many mucosa-associated disorders possess a specific gut microbiome fingerprint associated with the composition of the mucous barrier, which is further influenced by host-microbiome and inter-microbial interactions, dietary choices, microbe immigration and antimicrobials. Our review focuses on the interactions of diet (macronutrients and micronutrients), gut microbiota and mucous barriers (gastrointestinal and respiratory tract) and their importance in the onset and/or progression of major immune/metabolic disorders. We also highlight the key mechanisms that could be targeted therapeutically to prevent and/or treat these disorders.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades del Sistema Inmune , Microbiota , Dieta , Tracto Gastrointestinal , Humanos
10.
BMC Biol ; 16(1): 104, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30236109

RESUMEN

BACKGROUND: Defining the mechanisms that establish and regulate the transmission of epigenetic information from parent to offspring is critical for understanding disease heredity. Currently, the molecular pathways that regulate epigenetic information in the germline and its transmission to offspring are poorly understood. RESULTS: Here we provide evidence that Polycomb Repressive Complex 2 (PRC2) regulates paternal inheritance. Reduced PRC2 function in mice resulted in male sub-fertility and altered epigenetic and transcriptional control of retrotransposed elements in foetal male germ cells. Males with reduced PRC2 function produced offspring that over-expressed retrotransposed pseudogenes and had altered preimplantation embryo cleavage rates and cell cycle control. CONCLUSION: This study reveals a novel role for the histone-modifying complex, PRC2, in paternal intergenerational transmission of epigenetic effects on offspring, with important implications for understanding disease inheritance.


Asunto(s)
Epigénesis Genética/genética , Células Germinativas/metabolismo , Herencia Paterna/genética , Complejo Represivo Polycomb 2/genética , Animales , Masculino , Ratones , Complejo Represivo Polycomb 2/metabolismo
11.
Nucleic Acids Res ; 44(D1): D604-9, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26578596

RESUMEN

The Human Pan-Microbe Communities (HPMC) database (http://www.hpmcd.org/) provides a manually curated, searchable, metagenomic resource to facilitate investigation of human gastrointestinal microbiota. Over the past decade, the application of metagenome sequencing to elucidate the microbial composition and functional capacity present in the human microbiome has revolutionized many concepts in our basic biology. When sufficient high quality reference genomes are available, whole genome metagenomic sequencing can provide direct biological insights and high-resolution classification. The HPMC database provides species level, standardized phylogenetic classification of over 1800 human gastrointestinal metagenomic samples. This is achieved by combining a manually curated list of bacterial genomes from human faecal samples with over 21000 additional reference genomes representing bacteria, viruses, archaea and fungi with manually curated species classification and enhanced sample metadata annotation. A user-friendly, web-based interface provides the ability to search for (i) microbial groups associated with health or disease state, (ii) health or disease states and community structure associated with a microbial group, (iii) the enrichment of a microbial gene or sequence and (iv) enrichment of a functional annotation. The HPMC database enables detailed analysis of human microbial communities and supports research from basic microbiology and immunology to therapeutic development in human health and disease.


Asunto(s)
Bases de Datos de Ácidos Nucleicos , Genoma Microbiano , Metagenómica , Enfermedad , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/virología , Genes Microbianos , Humanos , Internet , Metagenómica/normas , Microbiota , Anotación de Secuencia Molecular , Estándares de Referencia , Análisis de Secuencia de ADN
12.
Nucleic Acids Res ; 43(2): 1177-88, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25539920

RESUMEN

Anti-microRNA (miRNA) oligonucleotides (AMOs) with 2'-O-Methyl (2'OMe) residues are commonly used to study miRNA function and can achieve high potency, with low cytotoxicity. Not withstanding this, we demonstrate the sequence-dependent capacity of 2'OMe AMOs to inhibit Toll-like receptor (TLR) 7 and 8 sensing of immunostimulatory RNA, independent of their miRNA-targeting function. Through a screen of 29 AMOs targeting common miRNAs, we found a subset of sequences highly inhibitory to TLR7 sensing in mouse macrophages. Interspecies conservation of this inhibitory activity was confirmed on TLR7/8 activity in human peripheral blood mononuclear cells. Significantly, we identified a core motif governing the inhibitory activity of these AMOs, which is present in more than 50 AMOs targeted to human miRNAs in miRBaseV20. DNA/locked nucleic acids (LNA) AMOs synthesized with a phosphorothioate backbone also inhibited TLR7 sensing in a sequence-dependent manner, demonstrating that the off-target effects of AMOs are not restricted to 2'OMe modification. Taken together, our work establishes the potential for off-target effects of AMOs on TLR7/8 function, which should be taken into account in their therapeutic development and in vivo application.


Asunto(s)
MicroARNs/antagonistas & inhibidores , Oligonucleótidos/química , Receptor Toll-Like 7/antagonistas & inhibidores , Receptor Toll-Like 8/antagonistas & inhibidores , Adyuvantes Inmunológicos/farmacología , Animales , Secuencia de Bases , Células HEK293 , Humanos , Ratones , Motivos de Nucleótidos , ARN/farmacología
13.
Bioinformatics ; 29(6): 810-2, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23396121

RESUMEN

Next-generation sequencing is rapidly becoming the approach of choice for transcriptional analysis experiments. Substantial advances have been achieved in computational approaches to support these technologies. These approaches typically rely on existing transcript annotations, introducing a bias towards known genes, require specific experimental design and computational resources, or focus only on identification of splice variants (ignoring other biologically relevant transcribed features contained within the data that may be important for downstream analysis). Biologically relevant transcribed features also include large and small non-coding RNA, new transcription start sites, alternative promoters, RNA editing and processing of coding transcripts. Also, many existing solutions lack accessible interfaces required for wide scale adoption. We present a user-friendly, rapid and computation-efficient feature annotation framework (RNA-eXpress) that enables identification of transcripts and other genomic and transcriptional features independently of current annotations. RNA-eXpress accepts mapped reads in the standard binary alignment (BAM) format and produces a study-specific feature annotation in GTF format, comparison statistics, sequence extraction and feature counts. The framework is designed to be easily accessible while allowing advanced users to integrate new feature-identification algorithms through simple class extension, thus facilitating expansion to novel feature types or identification of study-specific feature types.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Análisis de Secuencia de ARN/métodos , Programas Informáticos , Algoritmos , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Anotación de Secuencia Molecular , Isoformas de ARN/química , Empalme del ARN , ARN no Traducido/química , Sitio de Iniciación de la Transcripción , Regiones no Traducidas
14.
Sci Transl Med ; 15(678): eabq5126, 2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36630485

RESUMEN

Interactions between the developing microbiome and maturing immune system in early life are critical for establishment of a homeostasis beneficial to both host and commensals. The lung harbors a diverse community of microbes associated with health and local or systemic disease. We discuss how early life colonization and community changes correlate with immune development and health and disease throughout infancy, childhood, and adult life. We highlight key advances in microbiology, immunology, and computational biology that allow investigation of the functional relevance of interactions between the respiratory microbiome and host immune system, which may unlock the potential for microbiome-based therapeutics.


Asunto(s)
Microbiota , Pulmón/microbiología , Sistema Inmunológico
15.
Nat Commun ; 14(1): 6546, 2023 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-37863966

RESUMEN

Many gut microorganisms critical to human health rely on nutrients produced by each other for survival; however, these cross-feeding interactions are still challenging to quantify and remain poorly characterized. Here, we introduce a Metabolite Exchange Score (MES) to quantify those interactions. Using metabolic models of prokaryotic metagenome-assembled genomes from over 1600 individuals, MES allows us to identify and rank metabolic interactions that are significantly affected by a loss of cross-feeding partners in 10 out of 11 diseases. When applied to a Crohn's disease case-control study, our approach identifies a lack of species with the ability to consume hydrogen sulfide as the main distinguishing microbiome feature of disease. We propose that our conceptual framework will help prioritize in-depth analyses, experiments and clinical targets, and that targeting the restoration of microbial cross-feeding interactions is a promising mechanism-informed strategy to reconstruct a healthy gut ecosystem.


Asunto(s)
Enfermedad de Crohn , Microbioma Gastrointestinal , Microbiota , Humanos , Estudios de Casos y Controles , Metagenoma
16.
Cell Rep Med ; 4(7): 101124, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37467722

RESUMEN

Abnormal immune responses to the resident gut microbiome can drive inflammatory bowel disease (IBD). Here, we combine high-resolution, culture-based shotgun metagenomic sequencing and analysis with matched host transcriptomics across three intestinal sites (terminal ileum, cecum, rectum) from pediatric IBD (PIBD) patients (n = 58) and matched controls (n = 42) to investigate this relationship. Combining our site-specific approach with bacterial culturing, we establish a cohort-specific bacterial culture collection, comprising 6,620 isolates (170 distinct species, 32 putative novel), cultured from 286 mucosal biopsies. Phylogeny-based, clade-specific metagenomic analysis identifies key, functionally distinct Enterococcus clades associated with either IBD or health. Strain-specific in vitro validation demonstrates differences in cell cytotoxicity and inflammatory signaling in intestinal epithelial cells, consistent with the colonic mucosa-specific response measured in patients with IBD. This demonstrates the importance of strain-specific phenotypes and consideration of anatomical sites in exploring the dysregulated host-bacterial interactions in IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Humanos , Enfermedades Inflamatorias del Intestino/genética , Colon/patología , Biopsia , Mucosa Intestinal/microbiología , Células Epiteliales/patología
17.
Microb Genom ; 9(4)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37079454

RESUMEN

Globally, the anaerobic bacterium Clostridium perfringens causes severe disease in a wide array of hosts; however, C. perfringens strains are also carried asymptomatically. Accessory genes are responsible for much of the observed phenotypic variation and virulence within this species, with toxins frequently encoded on conjugative plasmids and many isolates carrying up to 10 plasmids. Despite this unusual biology, current genomic analyses have largely excluded isolates from healthy hosts or environmental sources. Accessory genomes, including plasmids, also have often been excluded from broader scale phylogenetic investigations. Here we interrogate a comprehensive collection of 464 C. perfringens genomes and identify the first putative non-conjugative enterotoxin (CPE)-encoding plasmids and a putative novel conjugative locus (Bcp) with sequence similarity to a locus reported from Clostridium botulinum. We sequenced and archived 102 new C. perfringens genomes, including those from rarely sequenced toxinotype B, C, D and E isolates. Long-read sequencing of 11 C. perfringens strains representing all toxinotypes (A-G) identified 55 plasmids from nine distinct plasmid groups. Interrogation of the 464 genomes in this collection identified 1045 plasmid-like contigs from the nine plasmid families, with a wide distribution across the C. perfringens isolates. Plasmids and plasmid diversity play an essential role in C. perfringens pathogenicity and broader biology. We have expanded the C. perfringens genome collection to include temporal, spatial and phenotypically diverse isolates including those carried asymptomatically in the gastrointestinal microbiome. This analysis has resulted in the identification of novel C. perfringens plasmids whilst providing a comprehensive understanding of species diversity.


Asunto(s)
Toxinas Bacterianas , Clostridium perfringens , Humanos , Toxinas Bacterianas/genética , Filogenia , Composición de Base , Análisis de Secuencia de ADN , ARN Ribosómico 16S , Plásmidos/genética
18.
Nat Commun ; 13(1): 1445, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35301310

RESUMEN

Mobile genetic elements (MGEs) carrying antibiotic resistance genes (ARGs) disseminate ARGs when they mobilise into new bacterial hosts. The nature of such horizontal gene transfer (HGT) events between human gut commensals and pathogens remain poorly characterised. Here, we compare 1354 cultured commensal strains (540 species) to 45,403 pathogen strains (12 species) and find 64,188 MGE-mediated ARG transfer events between the two groups using established methods. Among the 5931 MGEs, we find 15 broad host range elements predicted to have crossed different bacterial phyla while also occurring in animal and environmental microbiomes. We experimentally demonstrate that predicted broad host range MGEs can mobilise from commensals Dorea longicatena and Hungatella hathewayi to pathogen Klebsiella oxytoca, crossing phyla simultaneously. Our work establishes the MGE-mediated ARG dissemination network between human gut commensals and pathogens and highlights broad host range MGEs as targets for future ARG dissemination management.


Asunto(s)
Especificidad del Huésped , Microbiota , Animales , Antibacterianos/farmacología , Bacterias/genética , Farmacorresistencia Microbiana/genética , Genes Bacterianos , Especificidad del Huésped/genética , Humanos , Secuencias Repetitivas Esparcidas/genética , Microbiota/genética
19.
Aliment Pharmacol Ther ; 56(2): 192-208, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35611465

RESUMEN

BACKGROUND: From consumption of fermented foods and probiotics to emerging applications of faecal microbiota transplantation, the health benefit of manipulating the human microbiota has been exploited for millennia. Despite this history, recent technological advances are unlocking the capacity for targeted microbial manipulation as a novel therapeutic. AIM: This review summarises the current developments in microbiome-based medicines and provides insight into the next steps required for therapeutic development. METHODS: Here we review current and emerging approaches and assess the capabilities and weaknesses of these technologies to provide safe and effective clinical interventions. Key literature was identified through Pubmed searches with the following key words, 'microbiome', 'microbiome biomarkers', 'probiotics', 'prebiotics', 'synbiotics', 'faecal microbiota transplant', 'live biotherapeutics', 'microbiome mimetics' and 'postbiotics'. RESULTS: Improved understanding of the human microbiome and recent technological advances provide an opportunity to develop a new generation of therapies. These therapies will range from dietary interventions, prebiotic supplementations, single probiotic bacterial strains, human donor-derived faecal microbiota transplants, rationally selected combinations of bacterial strains as live biotherapeutics, and the beneficial products or effects produced by bacterial strains, termed microbiome mimetics. CONCLUSIONS: Although methods to identify and refine these therapeutics are continually advancing, the rapid emergence of these new approaches necessitates accepted technological and ethical frameworks for measurement, testing, laboratory practices and clinical translation.


Asunto(s)
Microbiota , Probióticos , Simbióticos , Trasplante de Microbiota Fecal , Humanos , Prebióticos , Probióticos/uso terapéutico
20.
Cell Host Microbe ; 30(1): 124-138.e8, 2022 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-34971560

RESUMEN

Human health and disease have increasingly been shown to be impacted by the gut microbiota, and mouse models are essential for investigating these effects. However, the compositions of human and mouse gut microbiotas are distinct, limiting translation of microbiota research between these hosts. To address this, we constructed the Mouse Gastrointestinal Bacteria Catalogue (MGBC), a repository of 26,640 high-quality mouse microbiota-derived bacterial genomes. This catalog enables species-level analyses for mapping functions of interest and identifying functionally equivalent taxa between the microbiotas of humans and mice. We have complemented this with a publicly deposited collection of 223 bacterial isolates, including 62 previously uncultured species, to facilitate experimental investigation of individual commensal bacteria functions in vitro and in vivo. Together, these resources provide the ability to identify and test functionally equivalent members of the host-specific gut microbiotas of humans and mice and support the informed use of mouse models in human microbiota research.


Asunto(s)
Bacterias/clasificación , Bacterias/aislamiento & purificación , Microbioma Gastrointestinal/fisiología , Animales , Bacterias/genética , Bacterias/metabolismo , Butiratos/metabolismo , Genoma Bacteriano , Humanos , Metagenoma/genética , Ratones , Modelos Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA