Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
2.
Cell Rep Med ; 5(3): 101426, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38366600

RESUMEN

The human gut microbiota is influenced by various factors, including health status and environmental conditions, yet considerable inter-individual differences remain unexplained. Previous studies identified that the gut microbiota of men who have sex with men (MSM) is distinct from that of non-MSM. Here, we reveal through species-level microbiota analysis using shotgun metagenomics that the gut microbiota of many MSM with Western origin resembles gut microbial communities of non-Westernized populations. Specifically, MSM gut microbiomes are frequently dominated by members of the Prevotellaceae family, including co-colonization of species from the Segatella copri complex and unknown Prevotellaceae members. Questionnaire-based analysis exploring inter-individual differences in MSM links specific sexual practices to microbiota composition. Moreover, machine learning identifies microbial features associated with sexual activities in MSM. Together, this study shows associations of sexual activities with gut microbiome alterations in MSM, which may have a large impact on population-based microbiota studies.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Minorías Sexuales y de Género , Masculino , Humanos , Homosexualidad Masculina , Conducta Sexual
3.
NPJ Precis Oncol ; 8(1): 10, 2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38200223

RESUMEN

The consensus molecular subtypes (CMS) of colorectal cancer (CRC) is the most widely-used gene expression-based classification and has contributed to a better understanding of disease heterogeneity and prognosis. Nevertheless, CMS intratumoral heterogeneity restricts its clinical application, stressing the necessity of further characterizing the composition and architecture of CRC. Here, we used Spatial Transcriptomics (ST) in combination with single-cell RNA sequencing (scRNA-seq) to decipher the spatially resolved cellular and molecular composition of CRC. In addition to mapping the intratumoral heterogeneity of CMS and their microenvironment, we identified cell communication events in the tumor-stroma interface of CMS2 carcinomas. This includes tumor growth-inhibiting as well as -activating signals, such as the potential regulation of the ETV4 transcriptional activity by DCN or the PLAU-PLAUR ligand-receptor interaction. Our study illustrates the potential of ST to resolve CRC molecular heterogeneity and thereby help advance personalized therapy.

4.
Cell Host Microbe ; 31(11): 1804-1819.e9, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37883976

RESUMEN

The Segatella copri (formerly Prevotella copri) complex (ScC) comprises taxa that are key members of the human gut microbiome. It was previously described to contain four distinct phylogenetic clades. Combining targeted isolation with large-scale metagenomic analysis, we defined 13 distinct Segatella copri-related species, expanding the ScC complex beyond four clades. Complete genome reconstruction of thirteen strains from seven species unveiled the presence of genetically diverse large circular extrachromosomal elements. These elements are consistently present in most ScC species, contributing to intra- and inter-species diversities. The nine species-level clades present in humans display striking differences in prevalence and intra-species genetic makeup across human populations. Based on a meta-analysis, we found reproducible associations between members of ScC and the male sex and positive correlations with lower visceral fat and favorable markers of cardiometabolic health. Our work uncovers genomic diversity within ScC, facilitating a better characterization of the human microbiome.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Humanos , Masculino , Microbioma Gastrointestinal/genética , Metagenoma , Filogenia , Prevotella , Femenino
5.
Cell Rep ; 42(6): 112549, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37245209

RESUMEN

Transfer of the gut microbiota from wild to laboratory mice alters the host's immune status and enhances resistance to infectious and metabolic diseases, but understanding of which microbes and how they promote host fitness is only emerging. Our analysis of metagenomic sequencing data reveals that Helicobacter spp. are enriched in wild compared with specific-pathogen-free (SPF) and conventionally housed mice, with multiple species commonly co-colonizing their hosts. We create laboratory mice harboring three non-SPF Helicobacter spp. to evaluate their effect on mucosal immunity and colonization resistance to the enteropathogen Citrobacter rodentium. Our experiments reveal that Helicobacter spp. interfere with C. rodentium colonization and attenuate C. rodentium-induced gut inflammation in wild-type (WT) mice, even preventing lethal infection in Rag2-/- SPF mice. Further analyses suggest that Helicobacter spp. interfere with tissue attachment of C. rodentium, putatively by reducing the availability of mucus-derived sugars. These results unveil pivotal protective functions of wild mouse microbiota constituents against intestinal infection.


Asunto(s)
Infecciones por Enterobacteriaceae , Microbioma Gastrointestinal , Microbiota , Animales , Ratones , Citrobacter rodentium , Inmunidad Adaptativa , Ratones Endogámicos C57BL
6.
Microb Genom ; 8(10)2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36190827

RESUMEN

Campylobacteriosis is still the most commonly reported zoonosis in the European Union causing gastrointestinal disease in humans. One of the most common sources for these food-borne infections is broiler meat. Interactions between Campylobacter (C.) jejuni and the intestinal microbiota might influence Campylobacter colonization in chickens. The aim of the present study was to gain further knowledge about exclusive interactions of the host microbiota with C. jejuni in Campylobacter-specific phage-free chickens under standardized conditions and special biosafety precautions.Therefore, 12 artificially infected (C. jejuni inoculum with a challenge dose of 7.64 log10 c.f.u.) and 12 control chickens of the breed Ross 308 were kept under special biosafety measures in an animal facility. At day 42 of life, microbiota studies were performed on samples of caecal digesta and mucus. No Campylobacter-specific phages were detected by real-time PCR analysis of caecal digesta of control or artificially infected chickens. Amplification of the 16S rRNA gene was performed within the hypervariable region V4 and subsequently sequenced with Illumina MiSeq platform. R (version 4.0.2) was used to compare the microbiota between C. jejuni-negative and C. jejuni-positive chickens. The factor chickens' infection status contributed significantly to the differences in microbial composition of mucosal samples, explaining 10.6 % of the microbiota variation (P=0.007) and in digesta samples, explaining 9.69 % of the microbiota variation (P=0.015). The strongest difference between C. jejuni-non-infected and C. jejuni-infected birds was observed for the family Peptococcaceae whose presence in C. jejuni-infected birds could not be demonstrated. Further, several genera of the family Ruminococcaceae appeared to be depressed in its abundance due to Campylobacter infection. A negative correlation was found between Christensenellaceae R-7 group and Campylobacter in C. jejuni-colonised chickens, both genera potentially competing for substrate. This makes Christensenellaceae R-7 group highly interesting for further studies that aim to find control options for Campylobacter infections and assess the relevance of this finding for chicken health and Campylobacter colonization.


Asunto(s)
Bacteriófagos , Infecciones por Campylobacter , Campylobacter jejuni , Campylobacter , Microbiota , Enfermedades de las Aves de Corral , Animales , Bacteriófagos/genética , Campylobacter/genética , Infecciones por Campylobacter/veterinaria , Campylobacter jejuni/genética , Pollos , Humanos , Membrana Mucosa , ARN Ribosómico 16S/genética
7.
Nat Commun ; 13(1): 3964, 2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35803930

RESUMEN

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide, and therapeutic options for advanced HCC are limited. Here, we observe that intestinal dysbiosis affects antitumor immune surveillance and drives liver disease progression towards cancer. Dysbiotic microbiota, as seen in Nlrp6-/- mice, induces a Toll-like receptor 4 dependent expansion of hepatic monocytic myeloid-derived suppressor cells (mMDSC) and suppression of T-cell abundance. This phenotype is transmissible via fecal microbiota transfer and reversible upon antibiotic treatment, pointing to the high plasticity of the tumor microenvironment. While loss of Akkermansia muciniphila correlates with mMDSC abundance, its reintroduction restores intestinal barrier function and strongly reduces liver inflammation and fibrosis. Cirrhosis patients display increased bacterial abundance in hepatic tissue, which induces pronounced transcriptional changes, including activation of fibro-inflammatory pathways as well as circuits mediating cancer immunosuppression. This study demonstrates that gut microbiota closely shapes the hepatic inflammatory microenvironment opening approaches for cancer prevention and therapy.


Asunto(s)
Carcinoma Hepatocelular , Microbioma Gastrointestinal , Neoplasias Hepáticas , Microbiota , Animales , Carcinoma Hepatocelular/metabolismo , Disbiosis/complicaciones , Neoplasias Hepáticas/metabolismo , Ratones , Microambiente Tumoral
8.
Cell Host Microbe ; 29(11): 1663-1679.e7, 2021 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-34610293

RESUMEN

Gut colonization with multidrug-resistant (MDR) bacteria enhances the risk of bloodstream infections in susceptible individuals. We demonstrate highly variable degrees of ex vivo colonization resistance against a carbapenem-resistant Klebsiella pneumoniae strain in human feces samples and subsequently isolate diverse K. oxytoca strains from protected donors. Several of these K. oxytoca strains reduce gut colonization of MDR K. pneumoniae strains in antibiotic-treated and gnotobiotic mouse models. Comparative analysis of K. oxytoca strains coupled with CRISPR-Cas9-mediated deletion of casA, a protein essential for utilization of selected beta-glucosides, identified competition for specific carbohydrates as key in promoting colonization resistance. In addition to direct competition between K. oxytoca and K. pneumoniae, cooperation with additional commensals is required to reestablish full colonization resistance and gut decolonization. Finally, humanized microbiota mice generated from K. pneumoniae-susceptible donors are protected by K. oxytoca administration, demonstrating the potential of commensal K. oxytoca strains as next-generation probiotics.


Asunto(s)
Metabolismo de los Hidratos de Carbono , Heces/microbiología , Tracto Gastrointestinal/microbiología , Klebsiella oxytoca/fisiología , Klebsiella pneumoniae/crecimiento & desarrollo , Interacciones Microbianas , Inmunidad Adaptativa , Adulto , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Niño , Farmacorresistencia Bacteriana Múltiple , Microbioma Gastrointestinal , Vida Libre de Gérmenes , Glucósidos/metabolismo , Humanos , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/microbiología , Klebsiella oxytoca/genética , Klebsiella oxytoca/aislamiento & purificación , Klebsiella pneumoniae/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL
9.
EMBO J ; 40(23): e108287, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34676563

RESUMEN

Prevotella copri is a prevalent inhabitant of the human gut and has been associated with plant-rich diet consumption and diverse health states. The underlying genetic basis of these associations remains enigmatic due to the lack of genetic tools. Here, we developed a novel versatile genetic toolbox for rapid and efficient genetic insertion and allelic exchange applicable to P. copri strains from multiple clades. Enabled by the genetic platform, we systematically investigated the specificity of polysaccharide utilization loci (PULs) and identified four highly conserved PULs for utilizing arabinan, pectic galactan, arabinoxylan, and inulin, respectively. Further genetic and functional analysis of arabinan utilization systems illustrate that P. copri has evolved two distinct types of arabinan-processing PULs (PULAra ) and that the type-II PULAra is significantly enriched in individuals consuming a vegan diet compared to other diets. In summary, this genetic toolbox will enable functional genetic studies for P. copri in future.


Asunto(s)
Dieta Vegetariana , Microbioma Gastrointestinal , Sitios Genéticos , Genoma Bacteriano , Polisacáridos/metabolismo , Prevotella/genética , Prevotella/metabolismo , Heces/microbiología , Humanos , Prevotella/clasificación , Prevotella/aislamiento & purificación
10.
Immunity ; 54(11): 2565-2577.e6, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34582747

RESUMEN

Key aspects of intestinal T cells, including their antigen specificity and their selection by the microbiota and other intestinal antigens, as well as the contribution of individual T cell clones to regulatory and effector functions, remain unresolved. Here we tracked adoptively transferred T cell populations to specify the interrelation of T cell receptor repertoire and the gut antigenic environment. We show that dominant TCRα clonotypes were shared between interferon-γ- and interleukin-17-producing but not regulatory Foxp3+ T cells. Identical TCRα clonotypes accumulated in the colon of different individuals, whereas antibiotics or defined colonization correlated with the expansion of distinct expanded T cell clonotypes. Our results demonstrate key aspects of intestinal CD4+ T cell activation and suggest that few microbial species exert a dominant effect on the intestinal T cell repertoire during colitis. We speculate that dominant proinflammatory T cell clones might provide a therapeutic target in human inflammatory bowel disease.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Colitis/etiología , Colitis/metabolismo , Microbioma Gastrointestinal/inmunología , Interacciones Huésped-Patógeno/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Traslado Adoptivo , Biomarcadores , Colitis/patología , Colitis/terapia , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Humanos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
11.
Front Immunol ; 12: 671331, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34566952

RESUMEN

The intestinal microbiota modulates IL-22 production in the intestine, including the induction of IL-22-producing CD4+ T helper cells. Which specific bacteria are responsible for the induction of these cells is less well understood. Here, we demonstrate through the use of novel gnotobiotic knock-in reporter mice that segmented filamentous bacteria (SFB), which are known for their ability to induce Th17 cells, also induce distinct IL-17A negative CD4+ T cell populations in the intestine. A subset of these cells instead produces IL-22 upon restimulation ex vivo and also during enteric infections. Furthermore, they produce a distinct set of cytokines compared to Th17 cells including the differential expression of IL-17F and IFN-γ. Importantly, genetic models demonstrate that these cells, presumably Th22 cells, develop independently of intestinal Th17 cells. Together, our data identifies that besides Th17, SFB also induces CD4+ T cell populations, which serve as immediate source of IL-22 during intestinal inflammation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Microbioma Gastrointestinal/inmunología , Interleucinas/inmunología , Células Th17/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Interleucinas/biosíntesis , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Salmonella typhi , Células Th17/metabolismo , Fiebre Tifoidea/inmunología , Fiebre Tifoidea/microbiología , Interleucina-22
12.
Nat Metab ; 3(9): 1228-1241, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34552267

RESUMEN

Primary sclerosing cholangitis (PSC) is a chronic cholestatic liver disease of unknown aetiology for which there are no approved therapeutic options. Patients with PSC display changes in gut microbiota and in bile acid (BA) composition; however, the contribution of these alterations to disease pathogenesis remains controversial. Here we identify a role for microbiota-dependent changes in BA synthesis that modulates PSC pathophysiology. In a genetic mouse model of PSC, we show that loss of microbiota-mediated negative feedback control of BA synthesis results in increased hepatic BA concentrations, disruption of bile duct barrier function and, consequently, fatal liver injury. We further show that these changes are dependent on decreased BA signalling to the farnesoid X receptor, which modulates the activity of the rate-limiting enzyme in BA synthesis, CYP7A1. Moreover, patients with advanced stages of PSC show suppressed BA synthesis as measured by serum C4 levels, which is associated with poor disease prognosis. Our preclinical data highlight the microbiota-dependent dynamics of BA metabolism in cholestatic liver disease, which could be important for future therapies targeting BA and gut microbiome interactions, and identify C4 as a potential biomarker to functionally stratify patients with PSC and predict disease outcomes.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colestasis/metabolismo , Microbioma Gastrointestinal , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Animales , Antibacterianos/administración & dosificación , Colangitis Esclerosante/metabolismo , Colangitis Esclerosante/patología , Humanos , Hígado/metabolismo , Ratones , Pronóstico , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
13.
Pathogens ; 10(8)2021 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-34451532

RESUMEN

Minimizing the clinical signs of Enterococcus cecorum infections causing enterococcal spondylitis in broiler herds is successful when initiated as metaphylaxis in the first week of life. Mechanistically, either the Enterococcus species present at that time are reduced by antibiotic treatment or antibiotic treatment might induce changes in intestinal microbiota composition with an indirect and subsequent influence. The aim of the present study was to examine the cecal microbiota of chickens after administering lincospectin or different additives to evaluate whether these additives have lincospectin-like effects on microbiota. Therefore, 157,400 broiler chickens were reared in four chicken houses (~40,000 birds each) on a broiler farm with history of enterococcal spondylitis. Each flock was treated either with lincospectin or water soluble esterified butyrins, Bacillus (B.) licheniformis or palm oil was added via drinking water during the first days of life. Ten birds per house were dissected at days 11, 20 and 33 of life and cecal microbiota were analyzed (16S rRNA gene sequencing). Lincospectin treatment elicited significant changes in the cecal microbiota composition until slaughter age. Among the tested additives, effects of B. licheniformis on cecal microbiota composition were most similar to those seen after the treatment with lincospectin at day 11.

14.
Nat Commun ; 12(1): 3999, 2021 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-34183670

RESUMEN

Type-III secretion systems (T3SSs) of the bacterial flagellum and the evolutionarily related injectisome are capable of translocating proteins with a remarkable speed of several thousand amino acids per second. Here, we investigate how T3SSs are able to transport proteins at such a high rate while preventing the leakage of small molecules. Our mutational and evolutionary analyses demonstrate that an ensemble of conserved methionine residues at the cytoplasmic side of the T3SS channel create a deformable gasket (M-gasket) around fast-moving substrates undergoing export. The unique physicochemical features of the M-gasket are crucial to preserve the membrane barrier, to accommodate local conformational changes during active secretion, and to maintain stability of the secretion pore in cooperation with a plug domain (R-plug) and a network of salt-bridges. The conservation of the M-gasket, R-plug, and salt-bridge network suggests a universal mechanism by which the membrane integrity is maintained during high-speed protein translocation in all T3SSs.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Transporte de Proteínas/fisiología , Salmonella typhimurium/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Proteínas Bacterianas/metabolismo , Membrana Celular/metabolismo , Membrana Celular/fisiología , Flagelos/metabolismo , Salmonella typhimurium/genética
15.
Cell Mol Immunol ; 18(5): 1211-1221, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33762684

RESUMEN

Intestinal Foxp3+ regulatory T cell (Treg) subsets are crucial players in tolerance to microbiota-derived and food-borne antigens, and compelling evidence suggests that the intestinal microbiota modulates their generation, functional specialization, and maintenance. Selected bacterial species and microbiota-derived metabolites, such as short-chain fatty acids (SCFAs), have been reported to promote Treg homeostasis in the intestinal lamina propria. Furthermore, gut-draining mesenteric lymph nodes (mLNs) are particularly efficient sites for the generation of peripherally induced Tregs (pTregs). Despite this knowledge, the direct role of the microbiota and their metabolites in the early stages of pTreg induction within mLNs is not fully elucidated. Here, using an adoptive transfer-based pTreg induction system, we demonstrate that neither transfer of a dysbiotic microbiota nor dietary SCFA supplementation modulated the pTreg induction capacity of mLNs. Even mice housed under germ-free (GF) conditions displayed equivalent pTreg induction within mLNs. Further molecular characterization of these de novo induced pTregs from mLNs by dissection of their transcriptomes and accessible chromatin regions revealed that the microbiota indeed has a limited impact and does not contribute to the initialization of the Treg-specific epigenetic landscape. Overall, our data suggest that the microbiota is dispensable for the early stages of pTreg induction within mLNs.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Ganglios Linfáticos/inmunología , Mesenterio/inmunología , Linfocitos T Reguladores/inmunología , Animales , Cromatina/metabolismo , Disbiosis/microbiología , Disbiosis/patología , Epigénesis Genética/efectos de los fármacos , Ácidos Grasos Volátiles/farmacología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Perfilación de la Expresión Génica , Ganglios Linfáticos/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T Reguladores/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Transcriptoma/genética
16.
Sci Immunol ; 6(56)2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33547052

RESUMEN

Interleukin-17A- (IL-17A) and IL-17F-producing CD4+ T helper cells (TH17 cells) are implicated in the development of chronic inflammatory diseases, such as multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). TH17 cells also orchestrate leukocyte invasion of the central nervous system (CNS) and subsequent tissue damage. However, the role of IL-17A and IL-17F as effector cytokines is still confused with the encephalitogenic function of the cells that produce these cytokines, namely, TH17 cells, fueling a long-standing debate in the neuroimmunology field. Here, we demonstrated that mice deficient for IL-17A/F lose their susceptibility to EAE, which correlated with an altered composition of their gut microbiota. However, loss of IL-17A/F in TH cells did not diminish their encephalitogenic capacity. Reconstitution of a wild-type-like intestinal microbiota or reintroduction of IL-17A specifically into the gut epithelium of IL-17A/F-deficient mice reestablished their susceptibility to EAE. Thus, our data demonstrated that IL-17A and IL-17F are not encephalitogenic mediators but rather modulators of intestinal homeostasis that indirectly alter CNS-directed autoimmunity.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Microbioma Gastrointestinal/inmunología , Interleucina-17/metabolismo , Esclerosis Múltiple/inmunología , Traslado Adoptivo , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/patología , Trasplante de Microbiota Fecal , Femenino , Humanos , Interleucina-17/genética , Masculino , Ratones , Ratones Noqueados , Esclerosis Múltiple/patología , Células Th17/inmunología , Células Th17/trasplante
17.
Int J Med Microbiol ; 311(2): 151472, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33461110

RESUMEN

Numerous studies have associated alterations in the gut microbiota composition with almost every known inflammatory disease. However, proving the biological relevance of distinct microbial signatures and linking specific microorganisms to host phenotypes, remains a considerable challenge. Correspondingly, increased abundance of members of Prevotella genus within microbial communities colonizing distinct mucosal surfaces has been found in individuals diagnosed with rheumatoid arthritis, periodontitis, metabolic disorders, and intestinal and vaginal dysbiosis. Still, the role of Prevotella spp. in the incidence of these diseases continues to be debated. For many years, poor understanding of Prevotella biology could be in large part attributed to the lack of experimental tools. However, in the recent years significant advances have been made towards overcoming these limitations, including increased number of isolates and improved understanding of genetic diversity. Besides discussing the most relevant associations between Prevotella spp. and inflammatory disorders, in the present review we examine the recent efforts to expand the Prevotella experimental "toolbox" and we highlight remaining experimental challenges that should advance future research and our understanding of Prevotella-host interplay.


Asunto(s)
Disbiosis , Microbioma Gastrointestinal , Interacciones Microbiota-Huesped , Inflamación/microbiología , Prevotella/metabolismo , Tracto Gastrointestinal , Humanos , Intestinos
18.
Mucosal Immunol ; 14(1): 113-124, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32433514

RESUMEN

Diverse microbial signatures within the intestinal microbiota have been associated with intestinal and systemic inflammatory diseases, but whether these candidate microbes actively modulate host phenotypes or passively expand within the altered microbial ecosystem is frequently not known. Here we demonstrate that colonization of mice with a member of the genus Prevotella, which has been previously associated to colitis in mice, exacerbates intestinal inflammation. Our analysis revealed that Prevotella intestinalis alters composition and function of the ecosystem resulting in a reduction of short-chain fatty acids, specifically acetate, and consequently a decrease in intestinal IL-18 levels during steady state. Supplementation of IL-18 to Prevotella-colonized mice was sufficient to reduce intestinal inflammation. Hence, we conclude that intestinal Prevotella colonization results in metabolic changes in the microbiota, which reduce IL-18 production and consequently exacerbate intestinal inflammation, and potential systemic autoimmunity.


Asunto(s)
Infecciones por Bacteroidaceae/inmunología , Infecciones por Bacteroidaceae/microbiología , Microbioma Gastrointestinal/inmunología , Interacciones Huésped-Patógeno/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Prevotella/inmunología , Inmunidad Adaptativa , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Metagenoma , Metagenómica/métodos , Ratones , Ratones Noqueados , Mucositis/etiología , Mucositis/metabolismo , Mucositis/patología
19.
Cell Mol Gastroenterol Hepatol ; 11(4): 909-933, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33189892

RESUMEN

BACKGROUND & AIMS: Acute liver failure (ALF) represents an unmet medical need in Western countries. Although the link between intestinal dysbiosis and chronic liver disease is well-established, there is little evidence for a functional role of gut-liver interaction during ALF. Here we hypothesized that intestinal dysbiosis may affect ALF. METHODS: To test this hypothesis, we assessed the association of proton pump inhibitor (PPI) or long-term antibiotics (ABx) intake, which have both been linked to intestinal dysbiosis, and occurrence of ALF in the 500,000 participants of the UK BioBank population-based cohort. For functional studies, male Nlrp6-/- mice were used as a dysbiotic mouse model and injected with a sublethal dose of acetaminophen (APAP) or lipopolysaccharide (LPS) to induce ALF. RESULTS: Multivariate Cox regression analyses revealed a significantly increased risk (odds ratio, 2.3-3) for developing ALF in UK BioBank participants with PPI or ABx. Similarly, dysbiotic Nlrp6-/- mice displayed exacerbated APAP- and LPS-induced liver injury, which was linked to significantly reduced gut and liver tissue microbiota diversity and correlated with increased intestinal permeability at baseline. Fecal microbiota transfer (FMT) from Nlrp6-/- mice into wild-type (WT) mice augmented liver injury on APAP treatment in recipient WT mice, resembling the inflammatory phenotype of Nlrp6-/- mice. Specifically, FMT skewed monocyte polarization in WT mice toward a Ly6Chi inflammatory phenotype, suggesting a critical function of these cells as sensors of gut-derived signals orchestrating the inflammatory response. CONCLUSIONS: Our data show an important yet unknown function of intestinal microbiota during ALF. Intestinal dysbiosis was transferrable to healthy WT mice via FMT and aggravated liver injury. Our study highlights intestinal microbiota as a targetable risk factor for ALF.


Asunto(s)
Acetaminofén/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Modelos Animales de Enfermedad , Disbiosis/complicaciones , Microbioma Gastrointestinal , Receptores de Superficie Celular/fisiología , Analgésicos no Narcóticos/toxicidad , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Permeabilidad
20.
Cell Host Microbe ; 28(6): 838-852.e6, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33113351

RESUMEN

Prevotella spp. are a dominant bacterial genus within the human gut. Multiple Prevotella spp. co-exist in some individuals, particularly those consuming plant-based diets. Additionally, Prevotella spp. exhibit variability in the utilization of diverse complex carbohydrates. To investigate the relationship between Prevotella competition and diet, we isolated Prevotella species from the mouse gut, analyzed their genomes and transcriptomes in vivo, and performed competition experiments between species in mice. Diverse dominant Prevotella species compete for similar metabolic niches in vivo, which is linked to the upregulation of specific polysaccharide utilization loci (PULs). Complex plant-derived polysaccharides are required for Prevotella spp. expansion, with arabinoxylans having a prominent impact on species abundance. The most dominant Prevotella species encodes a specific tandem-repeat trsusC/D PUL that enables arabinoxylan utilization and is conserved in human Prevotella copri strains, particularly among those consuming a vegan diet. These findings suggest that efficient (arabino)xylan-utilization is a factor contributing to Prevotella dominance.


Asunto(s)
Microbioma Gastrointestinal , Polisacáridos/metabolismo , Prevotella/crecimiento & desarrollo , Xilanos/metabolismo , Animales , ADN Bacteriano , Sitios Genéticos , Genoma Bacteriano , Glicósido Hidrolasas/genética , Glicosiltransferasas/genética , Humanos , Metagenómica , Ratones , Ratones Endogámicos C57BL , Filogenia , Prevotella/clasificación , Prevotella/aislamiento & purificación , ARN Ribosómico 16S , Transcriptoma , Veganos , Secuenciación Completa del Genoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...