Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Gut ; 72(6): 1081-1092, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36167663

RESUMO

OBJECTIVES: Inflammatory bowel disease (IBD) results from a combination of genetic predisposition, dysbiosis of the gut microbiota and environmental factors, leading to alterations in the gastrointestinal immune response and chronic inflammation. Caspase recruitment domain 9 (Card9), one of the IBD susceptibility genes, has been shown to protect against intestinal inflammation and fungal infection. However, the cell types and mechanisms involved in the CARD9 protective role against inflammation remain unknown. DESIGN: We used dextran sulfate sodium (DSS)-induced and adoptive transfer colitis models in total and conditional CARD9 knock-out mice to uncover which cell types play a role in the CARD9 protective phenotype. The impact of Card9 deletion on neutrophil function was assessed by an in vivo model of fungal infection and various functional assays, including endpoint dilution assay, apoptosis assay by flow cytometry, proteomics and real-time bioenergetic profile analysis (Seahorse). RESULTS: Lymphocytes are not intrinsically involved in the CARD9 protective role against colitis. CARD9 expression in neutrophils, but not in epithelial or CD11c+cells, protects against DSS-induced colitis. In the absence of CARD9, mitochondrial dysfunction increases mitochondrial reactive oxygen species production leading to the premature death of neutrophilsthrough apoptosis, especially in oxidative environment. The decreased functional neutrophils in tissues might explain the impaired containment of fungi and increased susceptibility to intestinal inflammation. CONCLUSION: These results provide new insight into the role of CARD9 in neutrophil mitochondrial function and its involvement in intestinal inflammation, paving the way for new therapeutic strategies targeting neutrophils.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Camundongos , Animais , Neutrófilos/metabolismo , Sobrevivência Celular , Colite/induzido quimicamente , Colite/prevenção & controle , Inflamação/metabolismo , Camundongos Knockout , Mitocôndrias/metabolismo , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Proteínas Adaptadoras de Sinalização CARD/metabolismo
2.
Gut ; 72(7): 1296-1307, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36270778

RESUMO

OBJECTIVE: The extent to which tryptophan (Trp) metabolism alterations explain or influence the outcome of inflammatory bowel diseases (IBDs) is still unclear. However, several Trp metabolism end-products are essential to intestinal homeostasis. Here, we investigated the role of metabolites from the kynurenine pathway. DESIGN: Targeted quantitative metabolomics was performed in two large human IBD cohorts (1069 patients with IBD). Dextran sodium sulphate-induced colitis experiments in mice were used to evaluate effects of identified metabolites. In vitro, ex vivo and in vivo experiments were used to decipher mechanisms involved. Effects on energy metabolism were evaluated by different methods including Single Cell mEtabolism by profiling Translation inHibition. RESULTS: In mice and humans, intestinal inflammation severity negatively correlates with the amount of xanthurenic (XANA) and kynurenic (KYNA) acids. Supplementation with XANA or KYNA decreases colitis severity through effects on intestinal epithelial cells and T cells, involving Aryl hydrocarbon Receptor (AhR) activation and the rewiring of cellular energy metabolism. Furthermore, direct modulation of the endogenous tryptophan metabolism, using the recombinant enzyme aminoadipate aminotransferase (AADAT), responsible for the generation of XANA and KYNA, was protective in rodent colitis models. CONCLUSION: Our study identified a new mechanism linking Trp metabolism to intestinal inflammation and IBD. Bringing back XANA and KYNA has protective effects involving AhR and the rewiring of the energy metabolism in intestinal epithelial cells and CD4+ T cells. This study paves the way for new therapeutic strategies aiming at pharmacologically correcting its alterations in IBD by manipulating the endogenous metabolic pathway with AADAT.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Humanos , Animais , Camundongos , Triptofano/metabolismo , Doenças Inflamatórias Intestinais/tratamento farmacológico , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/metabolismo , Intestinos , Inflamação
3.
Gut ; 70(6): 1174-1182, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33272977

RESUMO

Metabolic disorders represent a growing worldwide health challenge due to their dramatically increasing prevalence. The gut microbiota is a crucial actor that can interact with the host by the production of a diverse reservoir of metabolites, from exogenous dietary substrates or endogenous host compounds. Metabolic disorders are associated with alterations in the composition and function of the gut microbiota. Specific classes of microbiota-derived metabolites, notably bile acids, short-chain fatty acids, branched-chain amino acids, trimethylamine N-oxide, tryptophan and indole derivatives, have been implicated in the pathogenesis of metabolic disorders. This review aims to define the key classes of microbiota-derived metabolites that are altered in metabolic diseases and their role in pathogenesis. They represent potential biomarkers for early diagnosis and prognosis as well as promising targets for the development of novel therapeutic tools for metabolic disorders.


Assuntos
Ácidos e Sais Biliares/metabolismo , Microbioma Gastrointestinal/fisiologia , Doenças Metabólicas/metabolismo , Animais , Biomarcadores/metabolismo , Ácidos Graxos Voláteis/metabolismo , Interações entre Hospedeiro e Microrganismos , Humanos , Indóis/metabolismo , Doenças Metabólicas/diagnóstico , Doenças Metabólicas/microbiologia , Doenças Metabólicas/terapia , Metilaminas/metabolismo , Triptofano/metabolismo
4.
Clin Sci (Lond) ; 131(6): 471-485, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28093453

RESUMO

Recent studies suggest that colonization of colonic mucosa by pathogenic Escherichia coli could be involved in the development of colorectal cancer (CRC), especially through the production of genotoxins such as colibactin and/or by interfering with the DNA mismatch repair (MMR) pathway that leads to microsatellite instability (MSI). The present study, performed on 88 CRC patients, revealed a significant increase in E. coli colonization in the MSI CRC phenotype. In the same way, E. coli persistence and internalization were increased in vitro in MMR-deficient cells. Moreover, we demonstrated that colibactin-producing E. coli induce inhibition of the mutL homologue 1 (MLH1) MMR proteins, which could lead to genomic instability. However, colibactin-producing E. coli were more frequently identified in microsatellite stable (MSS) CRC. The present study suggests differences in the involvement of colibactin-producing E. coli in colorectal carcinogenesis according to the CRC phenotype. Further host-pathogen interactions studies should take into account CRC phenotypes.


Assuntos
Neoplasias Colorretais/microbiologia , Escherichia coli/isolamento & purificação , Instabilidade de Microssatélites , Adulto , Idoso , Idoso de 80 Anos ou mais , Transformação Celular Neoplásica/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/cirurgia , Reparo de Erro de Pareamento de DNA/genética , Enzimas Reparadoras do DNA/metabolismo , DNA de Neoplasias/genética , Escherichia coli/metabolismo , Feminino , Interações Hospedeiro-Patógeno/genética , Humanos , Mucosa Intestinal/microbiologia , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Peptídeos/metabolismo , Policetídeos/metabolismo , Estudos Prospectivos
5.
Gut ; 64(3): 428-37, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24898815

RESUMO

OBJECTIVE: Adherent-invasive Escherichia coli (AIEC) are abnormally predominant on Crohn's disease (CD) ileal mucosa. AIEC strains adhere to enterocytes via interaction between type 1 pili and CEACAM6 receptors abnormally expressed on CD ileal mucosa, leading to gut inflammation. We analysed whether epigenetic mechanisms are involved in the upregulation of CEACAM6 expression in intestinal epithelial cells (IECs). DESIGN: Methylation of CEACAM6 promoter was analysed using bisulfite sequencing and site-specific methylation by SnapShot. pCpGfree reporter system was used to analyse CEACAM6 promoter activity. Transgenic mice expressing human CEACAM6 fed either standard food or a low-methyl diet (LMD) were orally challenged with 10(9) AIEC LF82. After 3 days, gut-associated AIEC and proinflammatory cytokines were quantified. RESULTS: Analysis of CEACAM6 gene promoter revealed potentially methylated dinucleotide CpGs within HIF-1-responsive elements (HREs). Methylation levels of CpG within CEACAM6 promoter were inversely correlated with CEACAM6 expression in IEC expressing various levels of CEACAM6. We show the critical role of HRE methylation and transcription factor HIF-1 in the regulation of CEACAM6 gene in IEC. This was confirmed in transgenic mice expressing human CEACAM6 fed a LMD. LMD-dependent HRE demethylation led to abnormal gut expression of CEACAM6, favouring AIEC colonisation and subsequent inflammation. CONCLUSIONS: HRE hypomethylation in CEACAM6 promoter correlates with high expression in IEC. Our findings suggest that abnormal DNA methylation leading to CEACAM6 increased expression and AIEC-mediated gut inflammation can be related to changes in nutritional habits, such as low intake in methyl donor molecules, leading to abnormal epigenetic marks in mouse model mimicking CD susceptibility.


Assuntos
Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Doença de Crohn/etiologia , Dieta/efeitos adversos , Infecções por Escherichia coli/complicações , Proteínas Ligadas por GPI/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Animais , Antígenos CD/fisiologia , Células CACO-2 , Moléculas de Adesão Celular/fisiologia , Doença de Crohn/metabolismo , Doença de Crohn/microbiologia , Metilação de DNA , Epigênese Genética , Infecções por Escherichia coli/metabolismo , Proteínas Ligadas por GPI/fisiologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Camundongos , Camundongos Transgênicos
6.
Microbiome ; 10(1): 91, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35698210

RESUMO

BACKGROUND: Innate immunity genes have been reported to affect susceptibility to inflammatory bowel diseases (IBDs) and colitis in mice. Dectin-1, a receptor for fungal cell wall ß-glucans, has been clearly implicated in gut microbiota modulation and modification of the susceptibility to gut inflammation. Here, we explored the role of Dectin-1 and Dectin-2 (another receptor for fungal cell wall molecules) deficiency in intestinal inflammation. DESIGN: Susceptibility to dextran sodium sulfate (DSS)-induced colitis was assessed in wild-type, Dectin-1 knockout (KO), Dectin-2KO, and double Dectin-1KO and Dectin-2KO (D-1/2KO) mice. Inflammation severity, as well as bacterial and fungal microbiota compositions, was monitored. RESULTS: While deletion of Dectin-1 or Dectin-2 did not have a strong effect on DSS-induced colitis, double deletion of Dectin-1 and Dectin-2 significantly protected the mice from colitis. The protection was largely mediated by the gut microbiota, as demonstrated by fecal transfer experiments. Treatment of D-1/2KO mice with opportunistic fungal pathogens or antifungal agents did not affect the protection against gut inflammation, suggesting that the fungal microbiota had no role in the protective phenotype. Amplicon-based microbiota analysis of the fecal bacterial and fungal microbiota of D-1/2KO mice confirmed the absence of changes in the mycobiota but strong modification of the bacterial microbiota. We showed that bacteria from the Lachnospiraceae family were at least partly involved in this protection and that treatment with Blautia hansenii was enough to recapitulate the protection. CONCLUSIONS: Deletion of both the Dectin-1 and Dectin-2 receptors triggered a global shift in the microbial gut environment, affecting, surprisingly, mainly the bacterial population and driving protective effects in colitis. Members of the Lachnospiraceae family seem to play a central role in this protection. These findings provide new insights into the role of the Dectin receptors, which have been described to date as affecting only the fungal population, in intestinal physiopathology and in IBD. Video Abstract.


Assuntos
Colite , Microbioma Gastrointestinal , Micobioma , Animais , Bactérias/genética , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Inflamação , Lectinas Tipo C/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
7.
Gut Microbes ; 13(1): 1-18, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33769191

RESUMO

Crohn's disease (CD) is a chronic and disabling inflammatory disorder of the gut that is profoundly influenced by intestinal microbiota composition, host genetics and environmental factors. Several groups worldwide have described an imbalance of the gut microbiome composition, called dysbiosis, in CD patients, with an increase in Proteobacteria and Bacteroidetes and a decrease in Firmicutes. A high prevalence of adherent-invasive Escherichia coli (AIEC) pathobionts has been identified in the intestinal mucosa of CD patients. A significant loss in the bacteria that produce short-chain fatty acids (SCFAs) with anti-inflammatory properties, such as propionate, is also a consequence of dysbiosis in CD patients. Here, the AIEC reference strain LF82 was able to degrade propionate in the gut, which was sufficient to counteract the anti-inflammatory effect of propionate both in in vitro models and in mice with DSS-induced colitis. The consumption of propionate by AIEC pathobionts leads to an increase in TNF-α production by macrophages upon infection through the bacterial methyl-citrate pathway. To induce the protective effects of SCFAs on the inflamed gut, we used a G-protein-coupled receptor 43 agonist (GPR43 agonist) that is not metabolizable by intestinal bacteria. Interestingly, this agonist showed anti-inflammatory properties and decreased the severity of colitis in AIEC-infected mice, as assessed by an improvement in the disease activity index (DAI) and a decrease in AIEC pathobiont encroachment. Taken together, these results highlight the effectiveness of GPR43 agonist treatment in the control of gut inflammation and improved our understanding of the ability of AIEC to modulate propionate availability to create an infectious niche to its advantage.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/microbiologia , Doença de Crohn/microbiologia , Escherichia coli/metabolismo , Propionatos/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Animais , Anti-Inflamatórios não Esteroides/metabolismo , Aderência Bacteriana , Colite Ulcerativa/metabolismo , Citocinas/metabolismo , Disbiose/microbiologia , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/patogenicidade , Infecções por Escherichia coli/microbiologia , Ácidos Graxos Voláteis/metabolismo , Fezes/química , Fezes/microbiologia , Microbioma Gastrointestinal , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Humanos , Mucosa Intestinal/microbiologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Propionatos/farmacologia , Células RAW 264.7
8.
Cell Rep ; 36(1): 109332, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34233192

RESUMO

Gut interleukin-17A (IL-17)-producing γδ T cells are tissue-resident cells that are involved in both host defense and regulation of intestinal inflammation. However, factors that regulate their functions are poorly understood. In this study, we find that the gut microbiota represses IL-17 production by cecal γδ T cells. Treatment with vancomycin, a Gram-positive bacterium-targeting antibiotic, leads to decreased production of short-chain fatty acids (SCFAs) by the gut microbiota. Our data reveal that these microbiota-derived metabolites, particularly propionate, reduce IL-17 and IL-22 production by intestinal γδ T cells. Propionate acts directly on γδ T cells to inhibit their production of IL-17 in a histone deacetylase-dependent manner. Moreover, the production of IL-17 by human IL-17-producing γδ T cells from patients with inflammatory bowel disease (IBD) is regulated by propionate. These data contribute to a better understanding of the mechanisms regulating gut γδ T cell functions and offer therapeutic perspectives of these cells.


Assuntos
Ácidos Graxos Voláteis/farmacologia , Microbioma Gastrointestinal , Interleucina-17/biossíntese , Intestinos/citologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Adulto , Animais , Ceco/citologia , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Inibidores de Histona Desacetilases/farmacologia , Humanos , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/patologia , Interleucinas/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Vancomicina/farmacologia , Interleucina 22
9.
Gut Microbes ; 11(3): 364-380, 2020 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29494278

RESUMO

The pathogenesis of Crohn's disease (CD) is multifactorial and involves genetic susceptibility, environmental triggers and intestinal microbiota. Adherent-invasive Escherichia coli (AIEC) are flagellated bacteria more prevalent in CD patients than in healthy subjects and promote chronic intestinal inflammation. We aim at deciphering the role of flagella and flagellin modulation by intestinal conditions. AIEC flagellum expression is required for optimal adhesion to and invasion of intestinal epithelial cells. Interestingly, differential flagellin regulation was observed between commensal E. coli (HS) and AIEC (LF82) strains: flagellum expression by AIEC bacteria, in contrast to that of commensal E. coli, is enhanced under intestinal conditions (the presence of bile acids and mucins). Flagella are involved in the ability of the AIEC LF82 strain to cross a mucus layer in vitro and in vivo, conferring a selective advantage in penetrating the mucus layer and reaching the epithelial surface. In a CEABAC10 mouse model, a non-motile mutant (LF82-ΔfliC) exhibits reduced colonization that is restored by a dextran sodium sulfate treatment that alters mucus layer integrity. Moreover, a mutant that continuously secretes flagellin (LF82-ΔflgM) triggers a stronger inflammatory response than the wild-type strain, and the mutant's ability to colonize the CEABAC10 mouse model is decreased. Overexpression of flagellin in bacteria in contact with epithelial cells can be detrimental to their virulence by inducing acute inflammation that enhances AIEC clearance. AIEC pathobionts must finely modulate flagellum expression during the infection process, taking advantage of their specific virulence gene regulation to improve their adaptability and flexibility within the gut environment.


Assuntos
Aderência Bacteriana , Escherichia coli/fisiologia , Flagelos/metabolismo , Intestinos/microbiologia , Animais , Aderência Bacteriana/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células CACO-2 , Contagem de Colônia Microbiana , Doença de Crohn/microbiologia , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/patogenicidade , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Flagelos/genética , Flagelina/genética , Flagelina/metabolismo , Regulação Bacteriana da Expressão Gênica , Células HT29 , Humanos , Mucosa Intestinal/microbiologia , Intestinos/química , Camundongos , Camundongos Endogâmicos C57BL , Muco/microbiologia , Mutação , Fenótipo
10.
Sci Rep ; 10(1): 12922, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32737335

RESUMO

Deficiencies in methyl-donor molecules (folate, B12 vitamin), DNA methylation alteration and high prevalence of Adherent-Invasive Escherichia coli (AIEC) are frequently observed in Crohn's disease (CD) patients. AIEC bacteria adhere to the enterocytes through abnormally expressed carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) glycoprotein on host cells. This work aims at studying the relationship between methyl-donor molecules and AIEC-induced intestinal inflammatory response. CEABAC10 mice, a mouse model of CD, were fed a control or Methyl-donor Supplemented diet (MS diet). CEACAM6 promoter was hypermethylated in intestinal epithelial cells from mice fed an MS diet, which was associated with a significant decrease in CEACAM6 expression. Transcriptomic analysis revealed increased expression of anti-microbial peptides, increase in HSP70 gene family expression and a decreased expression of inflammatory marker Calprotectin upon MS diet, associated to a lower ability of AIEC bacteria to colonize gut mucosa. We observed in a cohort of CD patients that serum folate concentration was inversely correlated to Crohn's disease endoscopic index of severity and to fecal inflammatory markers. This study demonstrates that methyl-donor supplementation through the diet induces a specific intestinal micro-environment limiting pathobiont colonization of the gut. Clinicians may wish to consider methyl-donor supplementation for methyl-donor deficient CD patients.


Assuntos
Antígenos CD/biossíntese , Moléculas de Adesão Celular/biossíntese , Doença de Crohn , Metilação de DNA , Infecções por Escherichia coli , Escherichia coli/metabolismo , Alimentos Formulados , Proteínas Ligadas por GPI/biossíntese , Mucosa Intestinal , Regiões Promotoras Genéticas , Animais , Antígenos CD/genética , Aderência Bacteriana , Moléculas de Adesão Celular/genética , Doença de Crohn/dietoterapia , Doença de Crohn/genética , Doença de Crohn/metabolismo , Doença de Crohn/microbiologia , Modelos Animais de Doenças , Infecções por Escherichia coli/dietoterapia , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/patologia , Feminino , Proteínas Ligadas por GPI/genética , Regulação da Expressão Gênica , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Camundongos Transgênicos
11.
Cell Host Microbe ; 23(6): 716-724, 2018 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-29902437

RESUMO

The gut microbiota is a crucial actor in human physiology. Many of these effects are mediated by metabolites that are either produced by the microbes or derived from the transformation of environmental or host molecules. Among the array of metabolites at the interface between these microorganisms and the host is the essential aromatic amino acid tryptophan (Trp). In the gut, the three major Trp metabolism pathways leading to serotonin (5-hydroxytryptamine), kynurenine (Kyn), and indole derivatives are under the direct or indirect control of the microbiota. In this review, we gather the most recent advances concerning the central role of Trp metabolism in microbiota-host crosstalk in health and disease. Deciphering the complex equilibrium between these pathways will facilitate a better understanding of the pathogenesis of human diseases and open therapeutic opportunities.


Assuntos
Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/microbiologia , Triptofano/metabolismo , Transtorno Autístico/metabolismo , Transtorno Autístico/microbiologia , Colite/metabolismo , Doenças Transmissíveis/metabolismo , Doença de Crohn/metabolismo , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/fisiologia , Humanos , Indóis/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Cinurenina/metabolismo , Síndrome Metabólica/metabolismo , Síndrome Metabólica/microbiologia , Obesidade/metabolismo , Serotonina/metabolismo , Síndrome do Intestino Curto/metabolismo , Síndrome do Intestino Curto/microbiologia
12.
Microbiome ; 6(1): 152, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30172257

RESUMO

BACKGROUND: Host-microbe balance maintains intestinal homeostasis and strongly influences inflammatory conditions such as inflammatory bowel diseases (IBD). Here we focused on bacteria-fungi interactions and their implications on intestinal inflammation, a poorly understood area. METHODS: Dextran sodium sulfate (DSS)-induced colitis was assessed in mice treated with vancomycin (targeting gram-positive bacteria) or colistin (targeting Enterobacteriaceae) and supplemented with either Saccharomyces boulardii CNCM I-745 or Candida albicans. Inflammation severity as well as bacterial and fungal microbiota compositions was monitored. RESULTS: While S. boulardii improved DSS-induced colitis and C. albicans worsened it in untreated settings, antibiotic treatment strongly modified DSS susceptibility and effects of fungi on colitis. Vancomycin-treated mice were fully protected from colitis, while colistin-treated mice retained colitis phenotype but were not affected anymore by administration of fungi. Antibacterial treatments not only influenced bacterial populations but also had indirect effects on fungal microbiota. Correlations between bacterial and fungal relative abundance were dramatically decreased in colistin-treated mice compared to vancomycin-treated and control mice, suggesting that colistin-sensitive bacteria are involved in interactions with fungi. Restoration of the Enterobacteriaceae population by administrating colistin-resistant Escherichia coli reestablished both beneficial effects of S. boulardii and pathogenic effects of C. albicans on colitis severity. This effect was at least partly mediated by an improved gut colonization by fungi. CONCLUSIONS: Fungal colonization of the gut is affected by the Enterobacteriaceae population, indirectly modifying effects of mycobiome on the host. This finding provides new insights into the role of inter-kingdom functional interactions in intestinal physiopathology and potentially in IBD.


Assuntos
Candida albicans/fisiologia , Colite/microbiologia , Enterobacteriaceae/fisiologia , Saccharomyces boulardii/fisiologia , Animais , Antibiose , Anticorpos/administração & dosagem , Candida albicans/genética , Candida albicans/isolamento & purificação , Colite/tratamento farmacológico , Modelos Animais de Doenças , Enterobacteriaceae/classificação , Enterobacteriaceae/genética , Enterobacteriaceae/isolamento & purificação , Feminino , Microbioma Gastrointestinal , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Saccharomyces boulardii/genética , Saccharomyces boulardii/isolamento & purificação
13.
Cell Metab ; 28(5): 737-749.e4, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30057068

RESUMO

The extent to which microbiota alterations define or influence the outcome of metabolic diseases is still unclear, but the byproducts of microbiota metabolism are known to have an important role in mediating the host-microbiota interaction. Here, we identify that in both pre-clinical and clinical settings, metabolic syndrome is associated with the reduced capacity of the microbiota to metabolize tryptophan into derivatives that are able to activate the aryl hydrocarbon receptor. This alteration is not merely an effect of the disease as supplementation with AhR agonist or a Lactobacillus strain, with a high AhR ligand-production capacity, leads to improvement of both dietary- and genetic-induced metabolic impairments, particularly glucose dysmetabolism and liver steatosis, through improvement of intestinal barrier function and secretion of the incretin hormone GLP-1. These results highlight the role of gut microbiota-derived metabolites as a biomarker and as a basis for novel preventative or therapeutic interventions for metabolic disorders.


Assuntos
Microbioma Gastrointestinal , Síndrome Metabólica/metabolismo , Síndrome Metabólica/microbiologia , Receptores de Hidrocarboneto Arílico/metabolismo , Triptofano/metabolismo , Animais , Limosilactobacillus reuteri/metabolismo , Ligantes , Masculino , Síndrome Metabólica/tratamento farmacológico , Síndrome Metabólica/terapia , Camundongos , Camundongos Endogâmicos C57BL , Probióticos/uso terapêutico , Receptores de Hidrocarboneto Arílico/agonistas
14.
Sci Rep ; 6: 19032, 2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26742586

RESUMO

Recent advances have shown that the abnormal inflammatory response observed in CD involves an interplay among intestinal microbiota, host genetics and environmental factors. The escalating consumption of fat and sugar in Western countries parallels an increased incidence of CD during the latter 20(th) century. The impact of a HF/HS diet in mice was evaluated for the gut micro-inflammation, intestinal microbiota composition, function and selection of an E. coli population. The HF/HS diet created a specific inflammatory environment in the gut, correlated with intestinal mucosa dysbiosis characterized by an overgrowth of pro-inflammatory Proteobacteria such as E. coli, a decrease in protective bacteria, and a significantly decreased of SCFA concentrations. The expression of GPR43, a SCFA receptor was reduced in mice treated with a HF/HS diet and reduced in CD patients compared with controls. Interestingly, mice treated with an agonist of GPR43 were protected against DSS-induced colitis. Finally, the transplantation of feces from HF/HS treated mice to GF mice increased susceptibility to AIEC infection. Together, our results demonstrate that a Western diet could aggravate the inflammatory process and that the activation of the GPR43 receptor pathway could be used as a new strategy to treat CD patients.


Assuntos
Colite/microbiologia , Dieta Ocidental/efeitos adversos , Suscetibilidade a Doenças , Disbiose/etiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Receptores Acoplados a Proteínas G/genética , Animais , Aderência Bacteriana/efeitos dos fármacos , Benzenoacetamidas/farmacologia , Colite/induzido quimicamente , Colite/genética , Colite/prevenção & controle , Doença de Crohn/etiologia , Doença de Crohn/genética , Doença de Crohn/microbiologia , Doença de Crohn/prevenção & controle , Dieta Hiperlipídica/efeitos adversos , Sacarose Alimentar/efeitos adversos , Disbiose/genética , Disbiose/microbiologia , Disbiose/prevenção & controle , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Ácidos Graxos Voláteis/metabolismo , Transplante de Microbiota Fecal , Feminino , Regulação da Expressão Gênica , Interação Gene-Ambiente , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Dodecilsulfato de Sódio
16.
Biomed Res Int ; 2014: 567929, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25580435

RESUMO

A trillion of microorganisms colonize the mammalian intestine. Most of them have coevolved with the host in a symbiotic relationship and some of them have developed strategies to promote their replication in the presence of competing microbiota. Recent evidence suggests that perturbation of the microbial community favors the emergence of opportunistic pathogens, in particular adherent-invasive Escherichia coli (AIEC) that can increase incidence and severity of gut inflammation in the context of Crohn's disease (CD). This review will report the importance of AIEC as triggers of intestinal inflammation, focusing on their impact on epithelial barrier function and stimulation of mucosal inflammation. Beyond manipulation of immune response, restoration of gut microbiota as a new treatment option for CD patients will be discussed.


Assuntos
Doença de Crohn/microbiologia , Infecções por Escherichia coli/microbiologia , Interações Hospedeiro-Patógeno , Inflamação/microbiologia , Aderência Bacteriana , Doença de Crohn/terapia , Escherichia coli/patogenicidade , Infecções por Escherichia coli/terapia , Humanos , Inflamação/patologia , Inflamação/terapia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Microbiota
17.
PLoS One ; 8(3): e59386, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23516631

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) O157:H7 are responsible for repeated food-poisoning cases often caused by contaminated burgers. EHEC infection is predominantly a pediatric illness, which can lead to life-threatening diseases. Ruminants are the main natural reservoir for EHEC and food contamination almost always originates from faecal contamination. In beef meat products, primary bacterial contamination occurs at the dehiding stage of slaughtering. The extracellular matrix (ECM) is the most exposed part of the skeletal muscles in beef carcasses. Investigating the adhesion to the main muscle fibrous ECM proteins, insoluble fibronectin, collagen I, III and IV, laminin-α2 and elastin, results demonstrated that the preceding growth conditions had a great influence on subsequent bacterial attachment. In the tested experimental conditions, maximal adhesion to fibril-forming collagens I or III occurred at 25°C and pH 7. Once initially adhered, exposure to lower temperatures, as applied to meat during cutting and storage, or acidification, as in the course of post-mortem physiological modifications of muscle, had no effect on detachment, except at pHu. In addition, dense biofilm formation occurred on immobilized collagen I or III and was induced in growth medium supplemented with collagen I in solution. From this first comprehensive investigation of EHEC adhesion to ECM proteins with respect to muscle biology and meat processing, new research directions for the development of innovative practices to minimize the risk of meat contamination are further discussed.


Assuntos
Colágeno Tipo III/farmacologia , Colágeno Tipo I/farmacologia , Matriz Extracelular/metabolismo , Animais , Biofilmes/efeitos dos fármacos , Bovinos , Meios de Cultura/farmacologia , Elastina/metabolismo , Escherichia coli/metabolismo , Fibronectinas/metabolismo , Concentração de Íons de Hidrogênio , Laminina/metabolismo , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA