Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.085
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(5): 1206-1222.e16, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38428395

RESUMEN

Plasmids are extrachromosomal genetic elements that often encode fitness-enhancing features. However, many bacteria carry "cryptic" plasmids that do not confer clear beneficial functions. We identified one such cryptic plasmid, pBI143, which is ubiquitous across industrialized gut microbiomes and is 14 times as numerous as crAssphage, currently established as the most abundant extrachromosomal genetic element in the human gut. The majority of mutations in pBI143 accumulate in specific positions across thousands of metagenomes, indicating strong purifying selection. pBI143 is monoclonal in most individuals, likely due to the priority effect of the version first acquired, often from one's mother. pBI143 can transfer between Bacteroidales, and although it does not appear to impact bacterial host fitness in vivo, it can transiently acquire additional genetic content. We identified important practical applications of pBI143, including its use in identifying human fecal contamination and its potential as an alternative approach to track human colonic inflammatory states.


Asunto(s)
Bacterias , Tracto Gastrointestinal , Metagenoma , Plásmidos , Humanos , Bacterias/genética , Bacteroidetes/genética , Heces/microbiología , Plásmidos/genética
2.
Cell ; 185(20): 3705-3719.e14, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36179667

RESUMEN

The intestinal microbiota is an important modulator of graft-versus-host disease (GVHD), which often complicates allogeneic hematopoietic stem cell transplantation (allo-HSCT). Broad-spectrum antibiotics such as carbapenems increase the risk for intestinal GVHD, but mechanisms are not well understood. In this study, we found that treatment with meropenem, a commonly used carbapenem, aggravates colonic GVHD in mice via the expansion of Bacteroides thetaiotaomicron (BT). BT has a broad ability to degrade dietary polysaccharides and host mucin glycans. BT in meropenem-treated allogeneic mice demonstrated upregulated expression of enzymes involved in the degradation of mucin glycans. These mice also had thinning of the colonic mucus layer and decreased levels of xylose in colonic luminal contents. Interestingly, oral xylose supplementation significantly prevented thinning of the colonic mucus layer in meropenem-treated mice. Specific nutritional supplementation strategies, including xylose supplementation, may combat antibiotic-mediated microbiome injury to reduce the risk for intestinal GVHD in allo-HSCT patients.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bacteroides , Carbapenémicos/farmacología , Carbapenémicos/uso terapéutico , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/etiología , Meropenem , Ratones , Mucinas/metabolismo , Moco/metabolismo , Polisacáridos/metabolismo , Xilosa
3.
Cell ; 185(3): 513-529.e21, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35120663

RESUMEN

The human gut microbiota resides within a diverse chemical environment challenging our ability to understand the forces shaping this ecosystem. Here, we reveal that fitness of the Bacteroidales, the dominant order of bacteria in the human gut, is an emergent property of glycans and one specific metabolite, butyrate. Distinct sugars serve as strain-variable fitness switches activating context-dependent inhibitory functions of butyrate. Differential fitness effects of butyrate within the Bacteroides are mediated by species-level variation in Acyl-CoA thioesterase activity and nucleotide polymorphisms regulating an Acyl-CoA transferase. Using in vivo multi-omic profiles, we demonstrate Bacteroides fitness in the human gut is associated together, but not independently, with Acyl-CoA transferase expression and butyrate. Our data reveal that each strain of the Bacteroides exists within a unique fitness landscape based on the interaction of chemical components unpredictable by the effect of each part alone mediated by flexibility in the core genome.


Asunto(s)
Microbioma Gastrointestinal , Metaboloma , Polisacáridos/metabolismo , Acilcoenzima A/metabolismo , Secuencia de Aminoácidos , Aminoácidos de Cadena Ramificada/metabolismo , Bacteroidetes/efectos de los fármacos , Bacteroidetes/genética , Bacteroidetes/crecimiento & desarrollo , Butiratos/química , Butiratos/farmacología , Coenzima A Transferasas/química , Coenzima A Transferasas/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Variación Genética/efectos de los fármacos , Concentración de Iones de Hidrógeno , Metaboloma/efectos de los fármacos , Metaboloma/genética , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Especificidad de la Especie , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/genética , Transcripción Genética/efectos de los fármacos
4.
Cell ; 180(4): 717-728.e19, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-32084341

RESUMEN

Consumption of glucosinolates, pro-drug-like metabolites abundant in Brassica vegetables, has been associated with decreased risk of certain cancers. Gut microbiota have the ability to metabolize glucosinolates, generating chemopreventive isothiocyanates. Here, we identify a genetic and biochemical basis for activation of glucosinolates to isothiocyanates by Bacteroides thetaiotaomicron, a prominent gut commensal species. Using a genome-wide transposon insertion screen, we identified an operon required for glucosinolate metabolism in B. thetaiotaomicron. Expression of BT2159-BT2156 in a non-metabolizing relative, Bacteroides fragilis, resulted in gain of glucosinolate metabolism. We show that isothiocyanate formation requires the action of BT2158 and either BT2156 or BT2157 in vitro. Monocolonization of mice with mutant BtΔ2157 showed reduced isothiocyanate production in the gastrointestinal tract. These data provide insight into the mechanisms by which a common gut bacterium processes an important dietary nutrient.


Asunto(s)
Bacteroides thetaiotaomicron/metabolismo , Carbohidratos de la Dieta/metabolismo , Glucosinolatos/metabolismo , Intestinos/microbiología , Animales , Bacteroides thetaiotaomicron/genética , Bacteroides thetaiotaomicron/patogenicidad , Regulación Bacteriana de la Expresión Génica , Humanos , Masculino , Ratones , Operón , Simbiosis
5.
Cell ; 183(5): 1312-1324.e10, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33212011

RESUMEN

Interferon (IFN)-Is are crucial mediators of antiviral immunity and homeostatic immune system regulation. However, the source of IFN-I signaling under homeostatic conditions is unclear. We discovered that commensal microbes regulate the IFN-I response through induction of IFN-ß by colonic DCs. Moreover, the mechanism by which a specific commensal microbe induces IFN-ß was identified. Outer membrane (OM)-associated glycolipids of gut commensal microbes belonging to the Bacteroidetes phylum induce expression of IFN-ß. Using Bacteroides fragilis and its OM-associated polysaccharide A, we determined that IFN-ß expression was induced via TLR4-TRIF signaling. Antiviral activity of this purified microbial molecule against infection with either vesicular stomatitis virus (VSV) or influenza was demonstrated to be dependent on the induction of IFN-ß. In a murine VSV infection model, commensal-induced IFN-ß regulated natural resistance to virus infection. Due to the physiological importance of IFN-Is, discovery of an IFN-ß-inducing microbial molecule represents a potential approach for the treatment of some human diseases.


Asunto(s)
Inmunidad Innata , Microbiota , Virosis/microbiología , Animales , Bacteroides fragilis/fisiología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Colon/patología , Colon/virología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glucolípidos/metabolismo , Inmunidad Innata/efectos de los fármacos , Interferón beta/sangre , Interferón beta/metabolismo , Masculino , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Polisacáridos Bacterianos/farmacología , Receptor Toll-Like 4/metabolismo , Vesiculovirus/fisiología , Virosis/genética
6.
Cell ; 169(3): 538-546.e12, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28431251

RESUMEN

Applying synthetic biology to engineer gut-resident microbes provides new avenues to investigate microbe-host interactions, perform diagnostics, and deliver therapeutics. Here, we describe a platform for engineering Bacteroides, the most abundant genus in the Western microbiota, which includes a process for high-throughput strain modification. We have identified a novel phage promoter and translational tuning strategy and achieved an unprecedented level of expression that enables imaging of fluorescent-protein-expressing Bacteroides stably colonizing the mouse gut. A detailed characterization of the phage promoter has provided a set of constitutive promoters that span over four logs of strength without detectable fitness burden within the gut over 14 days. These promoters function predictably over a 1,000,000-fold expression range in phylogenetically diverse Bacteroides species. With these promoters, unique fluorescent signatures were encoded to allow differentiation of six species within the gut. Fluorescent protein-based differentiation of isogenic strains revealed that priority of gut colonization determines colonic crypt occupancy.


Asunto(s)
Bacteroides/clasificación , Bacteroides/genética , Microbioma Gastrointestinal , Ingeniería Genética , Animales , Bacteroides/aislamiento & purificación , Vida Libre de Gérmenes , Proteínas Fluorescentes Verdes/genética , Ratones , Regiones Promotoras Genéticas
7.
Cell ; 169(3): 547-558.e15, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28431252

RESUMEN

The gut microbiota is implicated in numerous aspects of health and disease, but dissecting these connections is challenging because genetic tools for gut anaerobes are limited. Inducible promoters are particularly valuable tools because these platforms allow real-time analysis of the contribution of microbiome gene products to community assembly, host physiology, and disease. We developed a panel of tunable expression platforms for the prominent genus Bacteroides in which gene expression is controlled by a synthetic inducer. In the absence of inducer, promoter activity is fully repressed; addition of inducer rapidly increases gene expression by four to five orders of magnitude. Because the inducer is absent in mice and their diets, Bacteroides gene expression inside the gut can be modulated by providing the inducer in drinking water. We use this system to measure the dynamic relationship between commensal sialidase activity and liberation of mucosal sialic acid, a receptor and nutrient for pathogens. VIDEO ABSTRACT.


Asunto(s)
Bacteroides/genética , Microbioma Gastrointestinal , Ingeniería Genética/métodos , Animales , Bacteroides/clasificación , Expresión Génica , Humanos , Ratones , Neuraminidasa/metabolismo , Regiones Promotoras Genéticas
8.
Cell ; 171(3): 655-667.e17, 2017 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-29053971

RESUMEN

The gut microbiota contributes to the development of normal immunity but, when dysregulated, can promote autoimmunity through various non-antigen-specific effects on pathogenic and regulatory lymphocytes. Here, we show that an integrase expressed by several species of the gut microbial genus Bacteroides encodes a low-avidity mimotope of the pancreatic ß cell autoantigen islet-specific glucose-6-phosphatase-catalytic-subunit-related protein (IGRP206-214). Studies in germ-free mice monocolonized with integrase-competent, integrase-deficient, and integrase-transgenic Bacteroides demonstrate that the microbial epitope promotes the recruitment of diabetogenic CD8+ T cells to the gut. There, these effectors suppress colitis by targeting microbial antigen-loaded, antigen-presenting cells in an integrin ß7-, perforin-, and major histocompatibility complex class I-dependent manner. Like their murine counterparts, human peripheral blood T cells also recognize Bacteroides integrase. These data suggest that gut microbial antigen-specific cytotoxic T cells may have therapeutic value in inflammatory bowel disease and unearth molecular mimicry as a novel mechanism by which the gut microbiota can regulate normal immune homeostasis. PAPERCLIP.


Asunto(s)
Autoantígenos/inmunología , Bacteroides/inmunología , Colitis/inmunología , Microbioma Gastrointestinal , Glucosa-6-Fosfatasa/inmunología , Adulto , Animales , Bacteroides/clasificación , Bacteroides/enzimología , Colitis/microbiología , Femenino , Glucosa-6-Fosfatasa/genética , Humanos , Tejido Linfoide/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Persona de Mediana Edad , Imitación Molecular , Linfocitos T/inmunología
9.
Cell ; 167(5): 1339-1353.e21, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27863247

RESUMEN

Despite the accepted health benefits of consuming dietary fiber, little is known about the mechanisms by which fiber deprivation impacts the gut microbiota and alters disease risk. Using a gnotobiotic mouse model, in which animals were colonized with a synthetic human gut microbiota composed of fully sequenced commensal bacteria, we elucidated the functional interactions between dietary fiber, the gut microbiota, and the colonic mucus barrier, which serves as a primary defense against enteric pathogens. We show that during chronic or intermittent dietary fiber deficiency, the gut microbiota resorts to host-secreted mucus glycoproteins as a nutrient source, leading to erosion of the colonic mucus barrier. Dietary fiber deprivation, together with a fiber-deprived, mucus-eroding microbiota, promotes greater epithelial access and lethal colitis by the mucosal pathogen, Citrobacter rodentium. Our work reveals intricate pathways linking diet, the gut microbiome, and intestinal barrier dysfunction, which could be exploited to improve health using dietary therapeutics.


Asunto(s)
Fibras de la Dieta/administración & dosificación , Microbioma Gastrointestinal , Mucosa Intestinal/microbiología , Animales , Citrobacter rodentium/fisiología , Colitis/microbiología , Colon/microbiología , Susceptibilidad a Enfermedades , Infecciones por Enterobacteriaceae/microbiología , Escherichia coli , Femenino , Vida Libre de Gérmenes , Humanos , Masculino , Ratones , Mucina 2/genética
10.
Immunity ; 50(3): 692-706.e7, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30824326

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a severe form of lung fibrosis with a high mortality rate. However, the etiology of IPF remains unknown. Here, we report that alterations in lung microbiota critically promote pulmonary fibrosis pathogenesis. We found that lung microbiota was dysregulated, and the dysregulated microbiota in turn induced production of interleukin-17B (IL-17B) during bleomycin-induced mouse lung fibrosis. Either lung-microbiota depletion or IL-17B deficiency ameliorated the disease progression. IL-17B cooperated with tumor necrosis factor-α to induce expression of neutrophil-recruiting genes and T helper 17 (Th17)-cell-promoting genes. Three pulmonary commensal microbes, which belong to the genera Bacteroides and Prevotella, were identified to promote fibrotic pathogenesis through IL-17R signaling. We further defined that the outer membrane vesicles (OMVs) that were derived from the identified commensal microbes induced IL-17B production through Toll-like receptor-Myd88 adaptor signaling. Together our data demonstrate that specific pulmonary symbiotic commensals can promote lung fibrosis by regulating a profibrotic inflammatory cytokine network.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/microbiología , Interleucina-17/metabolismo , Pulmón/metabolismo , Pulmón/microbiología , Microbiota/fisiología , Animales , Bacteroides/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , Neutrófilos/metabolismo , Prevotella/metabolismo , Transducción de Señal/fisiología , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
11.
EMBO J ; 42(2): e112372, 2023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36472247

RESUMEN

Protein synthesis is crucial for cell growth and survival yet one of the most energy-consuming cellular processes. How, then, do cells sustain protein synthesis under starvation conditions when energy is limited? To accelerate the translocation of mRNA-tRNAs through the ribosome, bacterial elongation factor G (EF-G) hydrolyzes energy-rich guanosine triphosphate (GTP) for every amino acid incorporated into a protein. Here, we identify an EF-G paralog-EF-G2-that supports translocation without hydrolyzing GTP in the gut commensal bacterium Bacteroides thetaiotaomicron. EF-G2's singular ability to sustain protein synthesis, albeit at slow rates, is crucial for bacterial gut colonization. EF-G2 is ~10-fold more abundant than canonical EF-G1 in bacteria harvested from murine ceca and, unlike EF-G1, specifically accumulates during carbon starvation. Moreover, we uncover a 26-residue region unique to EF-G2 that is essential for protein synthesis, EF-G2 dissociation from the ribosome, and responsible for the absence of GTPase activity. Our findings reveal how cells curb energy consumption while maintaining protein synthesis to advance fitness in nutrient-fluctuating environments.


Asunto(s)
Bacteroides , Factor G de Elongación Peptídica , Animales , Ratones , Bacteroides/genética , Bacteroides/metabolismo , Guanosina Trifosfato/metabolismo , Hidrólisis , Factor G de Elongación Peptídica/genética , Factor G de Elongación Peptídica/química , Ribosomas/metabolismo , ARN de Transferencia/metabolismo
12.
Proc Natl Acad Sci U S A ; 121(10): e2321910121, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38422018

RESUMEN

Bacteroidota are abundant members of the human gut microbiota that shape the enteric landscape by modulating host immunity and degrading dietary- and host-derived glycans. These processes are mediated in part by Outer Membrane Vesicles (OMVs). Here, we developed a high-throughput screen to identify genes required for OMV biogenesis and its regulation in Bacteroides thetaiotaomicron (Bt). We identified a family of Dual membrane-spanning anti-sigma factors (Dma) that control OMV biogenesis. We conducted molecular and multiomic analyses to demonstrate that deletion of Dma1, the founding member of the Dma family, modulates OMV production by controlling the activity of the ECF21 family sigma factor, Das1, and its downstream regulon. Dma1 has a previously uncharacterized domain organization that enables Dma1 to span both the inner and outer membrane of Bt. Phylogenetic analyses reveal that this common feature of the Dma family is restricted to the phylum Bacteroidota. This study provides mechanistic insights into the regulation of OMV biogenesis in human gut bacteria.


Asunto(s)
Bacteroides thetaiotaomicron , Humanos , Bacteroides thetaiotaomicron/genética , Factor sigma , Filogenia
13.
Proc Natl Acad Sci U S A ; 121(6): e2311323121, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38294941

RESUMEN

Microbiota-centric interventions are limited by our incomplete understanding of the gene functions of many of its constituent species. This applies in particular to small RNAs (sRNAs), which are emerging as important regulators in microbiota species yet tend to be missed by traditional functional genomics approaches. Here, we establish CRISPR interference (CRISPRi) in the abundant microbiota member Bacteroides thetaiotaomicron for genome-wide sRNA screens. By assessing the abundance of different protospacer-adjacent motifs, we identify the Prevotella bryantii B14 Cas12a as a suitable nuclease for CRISPR screens in these bacteria and generate an inducible Cas12a expression system. Using a luciferase reporter strain, we infer guide design rules and use this knowledge to assemble a computational pipeline for automated gRNA design. By subjecting the resulting guide library to a phenotypic screen, we uncover the sRNA BatR to increase susceptibility to bile salts through the regulation of genes involved in Bacteroides cell surface structure. Our study lays the groundwork for unlocking the genetic potential of these major human gut mutualists and, more generally, for identifying hidden functions of bacterial sRNAs.


Asunto(s)
Bacteroides thetaiotaomicron , ARN Pequeño no Traducido , Humanos , Bacteroides thetaiotaomicron/genética , ARN Guía de Sistemas CRISPR-Cas , Bilis , ARN Bacteriano/genética , ARN Pequeño no Traducido/genética
14.
Proc Natl Acad Sci U S A ; 121(28): e2318691121, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38968121

RESUMEN

Dietary lipids play an essential role in regulating the function of the gut microbiota and gastrointestinal tract, and these luminal interactions contribute to mediating host metabolism. Palmitic Acid Hydroxy Stearic Acids (PAHSAs) are a family of lipids with antidiabetic and anti-inflammatory properties, but whether the gut microbiota contributes to their beneficial effects on host metabolism is unknown. Here, we report that treating chow-fed female and male germ-free (GF) mice with PAHSAs improves glucose tolerance, but these effects are lost upon high fat diet (HFD) feeding. However, transfer of feces from PAHSA-treated, but not vehicle-treated, chow-fed conventional mice increases insulin sensitivity in HFD-fed GF mice. Thus, the gut microbiota is necessary for, and can transmit, the insulin-sensitizing effects of PAHSAs in HFD-fed GF male mice. Analyses of the cecal metagenome and lipidome of PAHSA-treated mice identified multiple lipid species that associate with the gut commensal Bacteroides thetaiotaomicron (Bt) and with insulin sensitivity resulting from PAHSA treatment. Supplementing live, and to some degree, heat-killed Bt to HFD-fed female mice prevented weight gain, reduced adiposity, improved glucose tolerance, fortified the colonic mucus barrier and reduced systemic inflammation compared to HFD-fed controls. These effects were not observed in HFD-fed male mice. Furthermore, ovariectomy partially reversed the beneficial Bt effects on host metabolism, indicating a role for sex hormones in mediating the Bt probiotic effects. Altogether, these studies highlight the fact that PAHSAs can modulate the gut microbiota and that the microbiota is necessary for the beneficial metabolic effects of PAHSAs in HFD-fed mice.


Asunto(s)
Dieta Alta en Grasa , Microbioma Gastrointestinal , Resistencia a la Insulina , Obesidad , Animales , Masculino , Femenino , Ratones , Microbioma Gastrointestinal/efectos de los fármacos , Obesidad/metabolismo , Obesidad/microbiología , Obesidad/etiología , Dieta Alta en Grasa/efectos adversos , Ratones Endogámicos C57BL , Ácidos Esteáricos/metabolismo , Ácido Palmítico/metabolismo , Heces/microbiología , Ratones Obesos
15.
EMBO J ; 41(22): e110712, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36254590

RESUMEN

Bone marrow (BM)-resident hematopoietic stem and progenitor cells (HSPCs) are often activated following bacterial insults to replenish the host hemato-immune system, but how they integrate the associated tissue damage signals to initiate distal tissue repair is largely unknown. Here, we show that acute gut inflammation expands HSPCs in the BM and directs them to inflamed mesenteric lymph nodes through GM-CSFR activation for further expansion and potential differentiation into Ly6C+ /G+ myeloid cells specialized in gut tissue repair. We identified this process to be mediated by Bacteroides, a commensal gram-negative bacteria that activates innate immune signaling. These findings establish cross-organ communication between the BM and distant inflamed sites, whereby a certain subset of multipotent progenitors is specified to respond to imminent hematopoietic demands and to alleviate inflammatory symptoms.


Asunto(s)
Células Madre Hematopoyéticas , Inflamación , Humanos , Células Madre Hematopoyéticas/fisiología , Inflamación/patología , Diferenciación Celular , Transducción de Señal , Células Mieloides/patología
16.
Proc Natl Acad Sci U S A ; 120(27): e2306314120, 2023 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-37364113

RESUMEN

Extracellular vesicles are produced in all three domains of life, and their biogenesis has common ancient origins in eukaryotes and archaea. Although bacterial vesicles were discovered several decades ago and multiple roles have been attributed to them, no mechanism has been established for vesicles biogenesis in bacteria. For this reason, there is a significant level of skepticism about the biological relevance of bacterial vesicles. Bacteroides thetaiotaomicron (Bt), a prominent member of the human intestinal microbiota, produces significant amounts of outer membrane vesicles (OMVs) which have been proposed to play key physiological roles. Here, we employed a dual marker system, consisting of outer membrane- and OMV-specific markers fused to fluorescent proteins to visualize OMV biogenesis by time-lapse microscopy. Furthermore, we performed comparative proteomic analyses to show that, in Bt, the OMV cargo is adapted for the optimal utilization of different polysaccharides. We also show that a negatively charged N-terminal motif acts as a signal for protein sorting into OMVs irrespective of the nutrient availability. Our results demonstrate that OMV production is the result of a highly regulated process in Bt.


Asunto(s)
Bacteroides thetaiotaomicron , Vesículas Extracelulares , Humanos , Proteómica , Vesículas Extracelulares/metabolismo , Bacteroides thetaiotaomicron/metabolismo , Dieta , Polisacáridos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo
17.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35145026

RESUMEN

Bacteroides thetaiotaomicron is a gut symbiont that inhabits the mucus layer and adheres to and metabolizes food particles, contributing to gut physiology and maturation. Although adhesion and biofilm formation could be key features for B. thetaiotaomicron stress resistance and gut colonization, little is known about the determinants of B. thetaiotaomicron biofilm formation. We previously showed that the B. thetaiotaomicron reference strain VPI-5482 is a poor in vitro biofilm former. Here, we demonstrated that bile, a gut-relevant environmental cue, triggers the formation of biofilm in many B. thetaiotaomicron isolates and common gut Bacteroidales species. We determined that bile-dependent biofilm formation involves the production of the DNase BT3563 or its homologs, degrading extracellular DNA (eDNA) in several B. thetaiotaomicron strains. Our study therefore shows that, although biofilm matrix eDNA provides a biofilm-promoting scaffold in many studied Firmicutes and Proteobacteria, BT3563-mediated eDNA degradation is required to form B. thetaiotaomicron biofilm in the presence of bile.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacteroides thetaiotaomicron/enzimología , Bilis/metabolismo , Biopelículas/crecimiento & desarrollo , Desoxirribonucleasas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Proteínas Bacterianas/genética , Bacteroides thetaiotaomicron/genética , Bacteroides thetaiotaomicron/fisiología , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Desoxirribonucleasas/genética , Regulación Enzimológica de la Expresión Génica/fisiología
18.
J Bacteriol ; 206(1): e0042623, 2024 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-38174933

RESUMEN

Bile acids (BAs) are cholesterol-derived molecules that aid in digestion and nutrient absorption, regulate host metabolic processes, and influence physiology of the gut microbiota. Both the host and its microbiome contribute to enzymatic modifications that shape the chemical diversity of BAs in the gut. Several bacterial species have been reported to conjugate standard amino acids to BAs, but it was not known if bacteria conjugate BAs to other amine classes. Here, we show that Bacteroides fragilis strain P207, isolated from a bacterial bloom in the J-pouch of a patient with ulcerative colitis pouchitis, conjugates standard amino acids and the neuroactive amines γ-aminobutyric acid (GABA) and tyramine to deoxycholic acid. We extended this analysis to other human gut isolates and identified species that are competent to conjugate GABA and tyramine to primary and secondary BAs, and further identified diverse BA-GABA and BA-tyramine amides in human stool. A longitudinal metabolomic analysis of J-pouch contents of the patient from whom B. fragilis P207 was isolated revealed highly reduced levels of secondary bile acids and a shifting BA amide profile before, during, and after onset of pouchitis, including temporal changes in several BA-GABA amides. Treatment of pouchitis with ciprofloxacin was associated with a marked reduction of nearly all BA amides in the J-pouch. Our study expands the known repertoire of conjugated bile acids produced by bacteria to include BA conjugates to GABA and tyramine and demonstrates that these molecules are present in the human gut. IMPORTANCE BAs are modified in multiple ways by host enzymes and the microbiota to produce a chemically diverse set of molecules that assist in the digestive process and impact many physiological functions. This study reports the discovery of bacterial species that conjugate the neuroactive amines, GABA and tyramine, to primary and secondary BAs. We further present evidence that BA-GABA and BA-tyramine conjugates are present in the human gut, and document a shifting BA-GABA profile in a human pouchitis patient before, during, and after inflammation and antibiotic treatment. GABA and tyramine are common metabolic products of the gut microbiota and potent neuroactive molecules. GABA- and tyramine-conjugated BAs may influence receptor-mediated regulatory mechanisms of humans and their gut microbes, and absorption of these molecules and their entry into enterohepatic circulation may impact host physiology at distal tissue sites. This study defines new conjugated bile acids in the human gut.


Asunto(s)
Ácidos y Sales Biliares , Reservoritis , Humanos , Aminoácidos , Ácido gamma-Aminobutírico , Aminas , Catálisis , Amidas
19.
Am J Physiol Gastrointest Liver Physiol ; 326(5): G607-G621, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38502145

RESUMEN

Fecal microbiota transplantation (FMT) is a promising therapy for inflammatory bowel disease (IBD) via rectifying gut microbiota. The aim of this study was to identify a mechanism of how specific bacteria-associated immune response contributes to alleviated colitis. Forty donors were divided into high (donor H) and low (donor L) groups according to the diversity and the abundance of Bacteroides and Faecalibacterium by 16S rRNA sequencing. FMT was performed on dextran sulfate sodium (DSS)-induced colitis in mice. Mice with colitis showed significant improvement in intestinal injury and immune imbalance after FMT with group donor H (P < 0.05). Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii were identified as targeted strains in donor feces by real-time PCR and droplet digital PCR. Mice with colitis were treated with mono- or dual-bacterial gavage therapy. Dual-bacterial therapy significantly ameliorated intestinal injury compared with mono-bacterial therapy (P < 0.05). Dual-bacterial therapy increased the M2/M1 macrophage polarization and improved the Th17/Treg imbalance and elevated IL-10 production by Tregs compared with the DSS group (P < 0.05). Metabolomics showed increased abundance of lecithin in the glycerophospholipid metabolism pathway. In conclusion, B. thetaiotaomicron and F. prausnitzii, as the key bacteria in donor feces, alleviate colitis in mice. The mechanism may involve increasing lecithin and regulating IL-10 production of intestinal Tregs.NEW & NOTEWORTHY We demonstrate that donors with high abundance of Bacteroides and Faecalibacterium ameliorate dextran sulfate sodium (DSS)-induced colitis in mice by fecal microbiota transplantation (FMT). The combination therapy of Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii is superior to mono-bacterial therapy in ameliorating colitis in mice, of which mechanism may involve promoting lecithin and inducing IL-10 production of intestinal Tregs.


Asunto(s)
Bacteroides thetaiotaomicron , Colitis , Faecalibacterium prausnitzii , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Animales , Colitis/terapia , Colitis/microbiología , Colitis/inducido químicamente , Colitis/inmunología , Ratones , Masculino , Humanos , Sulfato de Dextran , Ratones Endogámicos C57BL , Interleucina-10/metabolismo , Adulto , Femenino , Heces/microbiología , Modelos Animales de Enfermedad , Persona de Mediana Edad
20.
J Neuroinflammation ; 21(1): 120, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38715051

RESUMEN

BACKGROUND: The human gut microbiome (GM) is involved in inflammation and immune response regulation. Dysbiosis, an imbalance in this ecosystem, facilitates pathogenic invasion, disrupts immune equilibrium, and potentially triggers diseases including various human leucocyte antigen (HLA)-B27-associated autoinflammatory and autoimmune diseases such as inflammatory bowel disease (IBD) and spondyloarthropathy (SpA). This study assesses compositional and functional alterations of the GM in patients with HLA-B27-associated non-infectious anterior uveitis (AU) compared to healthy controls. METHODS: The gut metagenomes of 20 patients with HLA-B27-associated non-infectious AU, 21 age- and sex-matched HLA-B27-negative controls, and 6 HLA-B27-positive healthy controls without a history of AU were sequenced using the Illumina NovaSeq 6000 platform for whole metagenome shotgun sequencing. To identify taxonomic and functional features with significantly different relative abundances between groups and to identify associations with clinical metadata, the multivariate association by linear models (MaAsLin) R package was applied. RESULTS: Significantly higher levels of the Eubacterium ramulus species were found in HLA-B27-negative controls (p = 0.0085, Mann-Whitney U-test). No significant differences in microbial composition were observed at all other taxonomic levels. Functionally, the lipid IVA biosynthesis pathway was upregulated in patients (p < 0.0001, Mann-Whitney U-test). A subgroup analysis comparing patients with an active non-infectious AU to their age- and sex-matched HLA-B27-negative controls, showed an increase of the species Phocaeicola vulgatus in active AU (p = 0.0530, Mann-Whitney U-test). An additional analysis comparing AU patients to age- and sex-matched HLA-B27-positive controls, showed an increase of the species Bacteroides caccae in controls (p = 0.0022, Mann-Whitney U-test). CONCLUSION: In our cohort, non-infectious AU development is associated with compositional and functional alterations of the GM. Further research is needed to assess the causality of these associations, offering potentially novel therapeutic strategies.


Asunto(s)
Microbioma Gastrointestinal , Antígeno HLA-B27 , Uveítis Anterior , Humanos , Antígeno HLA-B27/genética , Antígeno HLA-B27/inmunología , Femenino , Masculino , Microbioma Gastrointestinal/fisiología , Persona de Mediana Edad , Uveítis Anterior/microbiología , Uveítis Anterior/inmunología , Adulto , Estudios de Casos y Controles , Anciano
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA