Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gut Microbes ; 16(1): 2295384, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38126163

RESUMEN

The anaerobic bacterium Fusobacterium nucleatum is significantly associated with human colorectal cancer (CRC) and is considered a significant contributor to the disease. The mechanisms underlying the promotion of intestinal tumor formation by F. nucleatum have only been partially uncovered. Here, we showed that F. nucleatum releases a metabolite into the microenvironment that strongly activates NF-κB in intestinal epithelial cells via the ALPK1/TIFA/TRAF6 pathway. Furthermore, we showed that the released molecule had the biological characteristics of ADP-heptose. We observed that F. nucleatum induction of this pathway increased the expression of the inflammatory cytokine IL-8 and two anti-apoptotic genes known to be implicated in CRC, BIRC3 and TNFAIP3. Finally, it promoted the survival of CRC cells and reduced 5-fluorouracil chemosensitivity in vitro. Taken together, our results emphasize the importance of the ALPK1/TIFA pathway in Fusobacterium induced-CRC pathogenesis, and identify the role of ADP-H in this process.


Asunto(s)
Neoplasias Colorrectales , Microbioma Gastrointestinal , Humanos , Fusobacterium nucleatum/metabolismo , Composición de Base , Filogenia , ARN Ribosómico 16S , Análisis de Secuencia de ADN , Neoplasias Colorrectales/patología , Heptosas/metabolismo , Microambiente Tumoral
2.
Gut Microbes ; 14(1): 2110639, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36036242

RESUMEN

The commensal bacteria that make up the gut microbiota impact the health of their host on multiple levels. In particular, the interactions taking place between the microbe-associated molecule patterns (MAMPs) and pattern recognition receptors (PRRs), expressed by intestinal epithelial cells (IECs), are crucial for maintaining intestinal homeostasis. While numerous studies showed that TLRs and NLRs are involved in the control of gut homeostasis by commensal bacteria, the role of additional innate immune receptors remains unclear. Here, we seek for novel MAMP-PRR interactions involved in the beneficial effect of the commensal bacterium Akkermansia muciniphila on intestinal homeostasis. We show that A. muciniphila strongly activates NF-κB in IECs by releasing one or more potent activating metabolites into the microenvironment. By using drugs, chemical and gene-editing tools, we found that the released metabolite(s) enter(s) epithelial cells and activate(s) NF-κB via an ALPK1, TIFA and TRAF6-dependent pathway. Furthermore, we show that the released molecule has the biological characteristics of the ALPK1 ligand ADP-heptose. Finally, we show that A. muciniphila induces the expression of the MUC2, BIRC3 and TNFAIP3 genes involved in the maintenance of the intestinal barrier function and that this process is dependent on TIFA. Altogether, our data strongly suggest that the commensal A. muciniphila promotes intestinal homeostasis by activating the ALPK1/TIFA/TRAF6 axis, an innate immune pathway exclusively described so far in the context of Gram-negative bacterial infections.


Asunto(s)
Microbioma Gastrointestinal , FN-kappa B , Adenosina Difosfato , Akkermansia , Heptosas , Inmunidad Innata , Factor 6 Asociado a Receptor de TNF , Verrucomicrobia
3.
Front Immunol ; 12: 742584, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35024040

RESUMEN

A causal correlation between the metabolic disorders associated with sugar intake and disruption of the gastrointestinal (GI) homeostasis has been suggested, but the underlying mechanisms remain unclear. To unravel these mechanisms, we investigated the effect of physiological amounts of fructose and glucose on barrier functions and inflammatory status in various regions of the GI tract and on the cecal microbiota composition. C57BL/6 mice were fed chow diet and given 15% glucose or 15% fructose in drinking water for 9 weeks. We monitored caloric intake, body weight, glucose intolerance, and adiposity. The intestinal paracellular permeability, cytokine, and tight junction protein expression were assessed in the jejunum, cecum, and colon. In the cecum, the microbiota composition was determined. Glucose-fed mice developed a marked increase in total adiposity, glucose intolerance, and paracellular permeability in the jejunum and cecum while fructose absorption did not affect any of these parameters. Fructose-fed mice displayed increased circulation levels of IL6. In the cecum, both glucose and fructose intake were associated with an increase in Il13, Ifnγ, and Tnfα mRNA and MLCK protein levels. To clarify the relationships between monosaccharides and barrier function, we measured the permeability of Caco-2 cell monolayers in response to IFNγ+TNFα in the presence of glucose or fructose. In vitro, IFNγ+TNFα-induced intestinal permeability increase was less pronounced in response to fructose than glucose. Mice treated with glucose showed an enrichment of Lachnospiracae and Desulfovibrionaceae while the fructose increased relative abundance of Lactobacillaceae. Correlations between pro-inflammatory cytokine gene expression and bacterial abundance highlighted the potential role of members of Desulfovibrio and Lachnospiraceae NK4A136 group genera in the inflammation observed in response to glucose intake. The increase in intestinal inflammation and circulating levels of IL6 in response to fructose was observed in the absence of intestinal permeability modification, suggesting that the intestinal permeability alteration does not precede the onset of metabolic outcome (low-grade inflammation, hyperglycemia) associated with chronic fructose consumption. The data also highlight the deleterious effects of glucose on gut barrier function along the GI tract and suggest that Desulfovibrionaceae and Lachnospiraceae play a key role in the onset of GI inflammation in response to glucose.


Asunto(s)
Fructosa/farmacología , Glucosa/farmacología , Mucosa Intestinal/efectos de los fármacos , Animales , Células CACO-2 , Ciego/metabolismo , Citocinas/sangre , ADN Bacteriano/genética , Disbiosis/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Humanos , Inflamación/sangre , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Masculino , Ratones Endogámicos C57BL , Quinasa de Cadena Ligera de Miosina/metabolismo , Permeabilidad/efectos de los fármacos
4.
Immunity ; 53(2): 398-416.e8, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32814028

RESUMEN

Paneth cells are the primary source of C-type lysozyme, a ß-1,4-N-acetylmuramoylhydrolase that enzymatically processes bacterial cell walls. Paneth cells are normally present in human cecum and ascending colon, but are rarely found in descending colon and rectum; Paneth cell metaplasia in this region and aberrant lysozyme production are hallmarks of inflammatory bowel disease (IBD) pathology. Here, we examined the impact of aberrant lysozyme production in colonic inflammation. Targeted disruption of Paneth cell lysozyme (Lyz1) protected mice from experimental colitis. Lyz1-deficiency diminished intestinal immune responses to bacterial molecular patterns and resulted in the expansion of lysozyme-sensitive mucolytic bacteria, including Ruminococcus gnavus, a Crohn's disease-associated pathobiont. Ectopic lysozyme production in colonic epithelium suppressed lysozyme-sensitive bacteria and exacerbated colitis. Transfer of R. gnavus into Lyz1-/- hosts elicited a type 2 immune response, causing epithelial reprograming and enhanced anti-colitogenic capacity. In contrast, in lysozyme-intact hosts, processed R. gnavus drove pro-inflammatory responses. Thus, Paneth cell lysozyme balances intestinal anti- and pro-inflammatory responses, with implications for IBD.


Asunto(s)
Clostridiales/inmunología , Colitis Ulcerosa/patología , Muramidasa/genética , Muramidasa/metabolismo , Células de Paneth/metabolismo , Animales , Clostridiales/genética , Colitis Ulcerosa/microbiología , Enfermedad de Crohn/patología , Femenino , Microbioma Gastrointestinal/genética , Células Caliciformes/citología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT6/genética
5.
Sci Rep ; 9(1): 8897, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31222022

RESUMEN

Owing to the growing recognition of the gut microbiota as a main partner of human health, we are expecting that the number of indications for fecal microbiota transplantation (FMT) will increase. Thus, there is an urgent need for standardization of the entire process of fecal transplant production. This study provides a complete standardized procedure to prepare and store live and ready-to-use transplants that meet the standard requirements of good practices to applied use in pharmaceutical industry. We show that, if time before transformation to transplants would exceed 24 hours, fresh samples should not be exposed to temperatures above 20 °C, and refrigeration at 4 °C can be a safe solution. Oxygen-free atmosphere was not necessary and simply removing air above collected samples was sufficient to preserve viability. Transplants prepared in maltodextrin-trehalose solutions, stored in a -80 °C standard freezer and then rapidly thawed at 37 °C, retained the best revivification potential as  proven by 16S rRNA profiles, metabolomic fingerprints, and flow cytometry assays over a 3-month observation period. Maltodextrin-trehalose containing cryoprotectants were also efficient in preserving viability of lyophilized transplants, either in their crude or purified form, an option that can be attractive for fecal transplant biobanking and oral formulation.


Asunto(s)
Trasplante de Microbiota Fecal , Heces , Guías como Asunto , Manejo de Especímenes/métodos , Crioprotectores , Humanos , Polisacáridos , Trehalosa
6.
Sci Rep ; 9(1): 643, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30679727

RESUMEN

The ligand activated transcription factor, aryl hydrocarbon receptor (AhR) emerged as a critical regulator of immune and metabolic processes in the gastrointestinal tract. In the gut, a main source of AhR ligands derives from commensal bacteria. However, many of the reported microbiota-derived ligands have been restricted to indolyl metabolites. Here, by screening commensal bacteria supernatants on an AhR reporter system expressed in human intestinal epithelial cell line (IEC), we found that the short chain fatty acid (SCFA) butyrate induced AhR activity and the transcription of AhR-dependent genes in IECs. We showed that AhR ligand antagonists reduced the effects of butyrate on IEC suggesting that butyrate could act as a ligand of AhR, which was supported by the nuclear translocation of AhR induced by butyrate and in silico structural modelling. In conclusion, our findings suggest that (i) butyrate activates AhR pathway and AhR-dependent genes in human intestinal epithelial cell-lines (ii) butyrate is a potential ligand for AhR which is an original mechanism of gene regulation by SCFA.


Asunto(s)
Butiratos/metabolismo , Mucosa Intestinal/citología , Receptores de Hidrocarburo de Aril/metabolismo , Células CACO-2 , Células HT29 , Humanos , Ligandos , Modelos Moleculares , Dominios Proteicos , Receptores de Hidrocarburo de Aril/química , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
7.
Sci Rep ; 8(1): 9742, 2018 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-29950699

RESUMEN

The intestinal microbiota contributes to the global wellbeing of their host by their fundamental role in the induction and maintenance of a healthy immune system. Commensal bacteria shape the mucosal immune system by influencing the proportion and the activation state of anti-inflammatory regulatory T cells (Treg) by metabolites that are still only partially unravelled. Microbiota members such as Clostridiales provide a transforming growth factor ß (TGFß)-rich environment that promotes the accumulation of Treg cells in the gut. The intestinal epithelial cells (IECs) take a central part in this process, as they are a major source of TGFß1 upon bacterial colonisation. In this study, we investigated which gut commensal bacteria were able to regulate the TGFB1 human promoter in IECs using supernatants from cultured bacteria. We reported that Firmicutes and Fusobacteria supernatants were the most potent TGFB1 modulators in HT-29 cells. Furthermore, we demonstrated that butyrate was the main metabolite in bacterial supernatants accounting for TGFß1 increase. This butyrate-driven effect was independent of the G-protein coupled receptors GPR41, GPR43 and GPR109a, the transporter MCT1 as well as the transcription factors NF-κB and AP-1 present on TGFB1 promoter. Interestingly, HDAC inhibitors were inducing a similar TGFB1 increase suggesting that butyrate acted through its HDAC inhibitor properties. Finally, our results showed that SP1 was the main transcription factor mediating the HDAC inhibitor effect of butyrate on TGFB1 expression. This is, to our knowledge, the first characterisation of the mechanisms underlying TGFB1 regulation in IEC by commensal bacteria derived butyrate.


Asunto(s)
Butiratos/metabolismo , Células Epiteliales/metabolismo , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Intestinos/citología , Factor de Transcripción Sp1/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Células HT29 , Humanos , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Factor de Crecimiento Transformador beta1/genética
8.
Front Immunol ; 9: 2838, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619249

RESUMEN

Commensal bacteria are crucial for the development and maintenance of a healthy immune system therefore contributing to the global well-being of their host. A wide variety of metabolites produced by commensal bacteria are influencing host health but the characterization of the multiple molecular mechanisms involved in host-microbiota interactions is still only partially unraveled. The intestinal epithelial cells (IECs) take a central part in the host-microbiota dialogue by inducing the first microbial-derived immune signals. Amongst the numerous effector molecules modulating the immune responses produced by IECs, indoleamine 2,3-dioxygenase-1 (IDO-1) is essential for gut homeostasis. IDO-1 expression is dependent on the microbiota and despites its central role, how the commensal bacteria impacts its expression is still unclear. Therefore, we investigated the impact of individual cultivable commensal bacteria on IDO-1 transcriptional expression and found that the short chain fatty acid (SCFA) butyrate was the main metabolite controlling IDO-1 expression in human primary IECs and IEC cell-lines. This butyrate-driven effect was independent of the G-protein coupled receptors GPR41, GPR43, and GPR109a and of the transcription factors SP1, AP1, and PPARγ for which binding sites were reported in the IDO-1 promoter. We demonstrated for the first time that butyrate represses IDO-1 expression by two distinct mechanisms. Firstly, butyrate decreases STAT1 expression leading to the inhibition of the IFNγ-dependent and phosphoSTAT1-driven transcription of IDO-1. In addition, we described a second mechanism by which butyrate impairs IDO-1 transcription in a STAT1-independent manner that could be attributed to its histone deacetylase (HDAC) inhibitor property. In conclusion, our results showed that IDO-1 expression is down-regulated by butyrate via a dual mechanism: the reduction of STAT1 level and the HDAC inhibitor property of SCFAs.


Asunto(s)
Bacterias , Ácido Butírico , Regulación hacia Abajo/inmunología , Células Epiteliales , Microbioma Gastrointestinal/inmunología , Regulación del Desarrollo de la Expresión Génica/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa , Mucosa Intestinal , Bacterias/inmunología , Bacterias/metabolismo , Ácido Butírico/inmunología , Ácido Butírico/metabolismo , Células CACO-2 , Células Epiteliales/enzimología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Femenino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Mucosa Intestinal/enzimología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Masculino , Persona de Mediana Edad , Receptores Acoplados a Proteínas G/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo
9.
Sci Rep ; 7: 43199, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28266623

RESUMEN

In healthy subjects, the intestinal microbiota interacts with the host's epithelium, regulating gene expression to the benefit of both, host and microbiota. The underlying mechanisms remain poorly understood, however. Although many gut bacteria are not yet cultured, constantly growing culture collections have been established. We selected 57 representative commensal bacterial strains to study bacteria-host interactions, focusing on PPARγ, a key nuclear receptor in colonocytes linking metabolism and inflammation to the microbiota. Conditioned media (CM) were harvested from anaerobic cultures and assessed for their ability to modulate PPARγ using a reporter cell line. Activation of PPARγ transcriptional activity was linked to the presence of butyrate and propionate, two of the main metabolites of intestinal bacteria. Interestingly, some stimulatory CMs were devoid of these metabolites. A Prevotella and an Atopobium strain were chosen for further study, and shown to up-regulate two PPARγ-target genes, ANGPTL4 and ADRP. The molecular mechanisms of these activations involved the phosphorylation of PPARγ through ERK1/2. The responsible metabolites were shown to be heat sensitive but markedly diverged in size, emphasizing the diversity of bioactive compounds found in the intestine. Here we describe different mechanisms by which single intestinal bacteria can directly impact their host's health through transcriptional regulation.


Asunto(s)
Bacterias Anaerobias/crecimiento & desarrollo , Células Epiteliales/fisiología , Microbioma Gastrointestinal , Regulación de la Expresión Génica , Mucosa Intestinal/fisiología , PPAR gamma/metabolismo , Procesamiento Proteico-Postraduccional , Proteína 4 Similar a la Angiopoyetina/metabolismo , Bacterias Anaerobias/metabolismo , Butiratos/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Medios de Cultivo Condicionados , Humanos , Sistema de Señalización de MAP Quinasas , Perilipina-2/metabolismo , Fosforilación , Propionatos/metabolismo
10.
Sci Rep ; 7: 40248, 2017 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-28091525

RESUMEN

The digestion of dietary fibers is a major function of the human intestinal microbiota. So far this function has been attributed to the microorganisms inhabiting the colon, and many studies have focused on this distal part of the gastrointestinal tract using easily accessible fecal material. However, microbial fermentations, supported by the presence of short-chain fatty acids, are suspected to occur in the upper small intestine, particularly in the ileum. Using a fosmid library from the human ileal mucosa, we screened 20,000 clones for their activities against carboxymethylcellulose and xylans chosen as models of the major plant cell wall (PCW) polysaccharides from dietary fibres. Eleven positive clones revealed a broad range of CAZyme encoding genes from Bacteroides and Clostridiales species, as well as Polysaccharide Utilization Loci (PULs). The functional glycoside hydrolase genes were identified, and oligosaccharide break-down products examined from different polysaccharides including mixed-linkage ß-glucans. CAZymes and PULs were also examined for their prevalence in human gut microbiome. Several clusters of genes of low prevalence in fecal microbiome suggested they belong to unidentified strains rather specifically established upstream the colon, in the ileum. Thus, the ileal mucosa-associated microbiota encompasses the enzymatic potential for PCW polysaccharide degradation in the small intestine.


Asunto(s)
Fibras de la Dieta/metabolismo , Fibras de la Dieta/microbiología , Microbioma Gastrointestinal , Íleon/microbiología , Bacteroides/metabolismo , Metabolismo de los Hidratos de Carbono , Carboximetilcelulosa de Sodio/metabolismo , Mapeo Cromosómico , Clostridiales/metabolismo , Heces/microbiología , Humanos , Metagenoma , Metagenómica , Xilanos/metabolismo
11.
PLoS One ; 7(12): e52869, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23300800

RESUMEN

The human intestine is a balanced ecosystem well suited for bacterial survival, colonization and growth, which has evolved to be beneficial both for the host and the commensal bacteria. Here, we investigated the effect of bacterial metabolites produced by commensal bacteria on AP-1 signaling pathway, which has a plethora of effects on host physiology. Using intestinal epithelial cell lines, HT-29 and Caco-2, stably transfected with AP-1-dependent luciferase reporter gene, we tested the effect of culture supernatant from 49 commensal strains. We observed that several bacteria were able to activate the AP-1 pathway and this was correlated to the amount of short chain fatty acids (SCFAs) produced. Besides being a major source of energy for epithelial cells, SCFAs have been shown to regulate several signaling pathways in these cells. We show that propionate and butyrate are potent activators of the AP-1 pathway, butyrate being the more efficient of the two. We also observed a strong synergistic activation of AP-1 pathway when using butyrate with PMA, a PKC activator. Moreover, butyrate enhanced the PMA-induced expression of c-fos and ERK1/2 phosphorylation, but not p38 and JNK. In conclusion, we showed that SCFAs especially butyrate regulate the AP-1 signaling pathway, a feature that may contribute to the physiological impact of the gut microbiota on the host. Our results provide support for the involvement of butyrate in modulating the action of PKC in colon cancer cells.


Asunto(s)
Butiratos/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/citología , Acetato de Tetradecanoilforbol/farmacología , Factor de Transcripción AP-1/metabolismo , Antígenos de Diferenciación/metabolismo , Butiratos/farmacología , Células CACO-2 , Diferenciación Celular , Proliferación Celular , Medios de Cultivo Condicionados , Ciclina D1/genética , Ciclina D1/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ácidos Grasos/metabolismo , Ácidos Grasos/farmacología , Expresión Génica , Regulación de la Expresión Génica , Genes Reporteros , Células HT29 , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Mucosa Intestinal/microbiología , Luciferasas/biosíntesis , Luciferasas/genética , Sistema de Señalización de MAP Quinasas , Metagenoma , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factor de Transcripción AP-1/fisiología , Activación Transcripcional
12.
J Biomed Biotechnol ; 2011: 282356, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21765633

RESUMEN

The intestinal microbiota plays an important role in modulation of mucosal immune responses. To seek interactions between intestinal epithelial cells (IEC) and commensal bacteria, we screened 49 commensal strains for their capacity to modulate NF-κB. We used HT-29/kb-seap-25 and Caco-2/kb-seap-7 intestinal epithelial cells and monocyte-like THP-1 blue reporter cells to measure effects of commensal bacteria on cellular expression of a reporter system for NF-κB. Bacteria conditioned media (CM) were tested alone or together with an activator of NF-κB to explore its inhibitory potentials. CM from 8 or 10 different commensal species activated NF-κB expression on HT-29 and Caco-2 cells, respectively. On THP-1, CM from all but 5 commensal strains stimulated NF-κB. Upon challenge with TNF-α or IL-1ß, some CM prevented induced NF-κB activation, whereas others enhanced it. Interestingly, the enhancing effect of some CM was correlated with the presence of butyrate and propionate. Characterization of the effects of the identified bacteria and their implications in human health awaits further investigations.


Asunto(s)
Bacterias/química , Bacterias/metabolismo , Intestinos/inmunología , Intestinos/microbiología , FN-kappa B/metabolismo , Células CACO-2 , Técnicas de Cultivo de Célula , Medios de Cultivo Condicionados , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Expresión Génica/genética , Células HT29 , Humanos , Interleucina-1beta/química , Interleucina-1beta/metabolismo , Mucosa Intestinal/metabolismo , Monocitos , FN-kappa B/análisis , FN-kappa B/genética , Factor de Necrosis Tumoral alfa/química , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...