Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Gut Microbes ; 16(1): 2310215, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38374654

RESUMEN

Human colorectal cancers (CRCs) are readily colonized by colibactin-producing E. coli (CoPEC). CoPEC induces DNA double-strand breaks, DNA mutations, genomic instability, and cellular senescence. Infected cells produce a senescence-associated secretory phenotype (SASP), which is involved in the increase in tumorigenesis observed in CRC mouse models infected with CoPEC. This study investigated whether CoPEC, and the SASP derived from CoPEC-infected cells, impacted chemotherapeutic resistance. Human intestinal epithelial cells were infected with the CoPEC clinical 11G5 strain or with its isogenic mutant, which is unable to produce colibactin. Chemotherapeutic resistance was assessed in vitro and in a xenograft mouse model. Expressions of cancer stem cell (CSC) markers in infected cells were investigated. Data were validated using a CRC mouse model and human clinical samples. Both 11G5-infected cells, and uninfected cells incubated with the SASP produced by 11G5-infected cells exhibited an increased resistance to chemotherapeutic drugs in vitro and in vivo. This finding correlated with the induction of the epithelial to mesenchymal transition (EMT), which led to the emergence of cells exhibiting CSC features. They grew on ultra-low attachment plates, formed colonies in soft agar, and overexpressed several CSC markers (e.g. CD133, OCT-3/4, and NANOG). In agreement with these results, murine and human CRC biopsies colonized with CoPEC exhibited higher expression levels of OCT-3/4 and NANOG than biopsies devoid of CoPEC. Conclusion: CoPEC might aggravate CRCs by inducing the emergence of cancer stem cells that are highly resistant to chemotherapy.


Asunto(s)
Microbioma Gastrointestinal , Neoplasias , Péptidos , Policétidos , Humanos , Ratones , Animales , Escherichia coli/genética , Escherichia coli/metabolismo , Transición Epitelial-Mesenquimal , Mutágenos/metabolismo , Policétidos/farmacología , Policétidos/metabolismo , Modelos Animales de Enfermedad , Células Madre Neoplásicas/metabolismo
2.
Int J Mol Sci ; 22(7)2021 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-33805299

RESUMEN

BACKGROUND: Adherent-invasive Escherichia coli (AIEC) have been implicated in the etiology of Crohn's disease. The AIEC reference strain LF82 possesses a pathogenicity island similar to the high pathogenicity island of Yersinia spp., which encodes the yersiniabactin siderophore required for iron uptake and growth of the bacteria in iron-restricted environment. Here, we investigated the role of yersiniabactin during AIEC infection. METHODS: Intestinal epithelial T84 cells and CEABAC10 transgenic mice were infected with LF82 or its mutants deficient in yersiniabactin expression. Autophagy was assessed by Western blot analysis for p62 and LC3-II expression. RESULTS: Loss of yersiniabactin decreased the growth of LF82 in competitive conditions, reducing the ability of LF82 to adhere to and invade T84 cells and to colonize the intestinal tract of CEABAC10 mice. However, yersiniabactin deficiency increased LF82 intracellular replication. Mechanistically, a functional yersiniabactin is necessary for LF82-induced expression of HIF-1α, which is implicated in autophagy activation in infected cells. CONCLUSION: Our study highlights a novel role for yersiniabactin siderophore in AIEC-host interaction. Indeed, yersiniabactin, which is an advantage for AIEC to growth in a competitive environment, could be a disadvantage for the bacteria as it activates autophagy, a key host defense mechanism, leading to bacterial clearance.


Asunto(s)
Autofagia , Enfermedad de Crohn/etiología , Infecciones por Escherichia coli/complicaciones , Escherichia coli/patogenicidad , Mucosa Intestinal/fisiopatología , Fenoles/metabolismo , Tiazoles/metabolismo , Animales , Enfermedad de Crohn/fisiopatología , Escherichia coli/metabolismo , Infecciones por Escherichia coli/fisiopatología , Masculino , Ratones , Ratones Transgénicos
3.
Gut Microbes ; 13(1): 1-18, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33769191

RESUMEN

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.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/microbiología , Enfermedad de Crohn/microbiología , Escherichia coli/metabolismo , Propionatos/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Animales , Antiinflamatorios no Esteroideos/metabolismo , Adhesión Bacteriana , Colitis Ulcerosa/metabolismo , Citocinas/metabolismo , Disbiosis/microbiología , Escherichia coli/crecimiento & desarrollo , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/microbiología , Ácidos Grasos Volátiles/metabolismo , Heces/química , Heces/microbiología , Microbioma Gastrointestinal , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Humanos , Mucosa Intestinal/microbiología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Propionatos/farmacología , Células RAW 264.7
4.
Sci Rep ; 9(1): 2175, 2019 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-30778122

RESUMEN

The adherent-invasive Escherichia coli (AIEC), which colonize the ileal mucosa of Crohn's disease patients, adhere to intestinal epithelial cells, invade them and exacerbate intestinal inflammation. The high nutrient competition between the commensal microbiota and AIEC pathobiont requires the latter to occupy their own metabolic niches to survive and proliferate within the gut. In this study, a global RNA sequencing of AIEC strain LF82 has been used to observe the impact of bile salts on the expression of metabolic genes. The results showed a global up-regulation of genes involved in degradation and a down-regulation of those implicated in biosynthesis. The main up-regulated degradation pathways were ethanolamine, 1,2-propanediol and citrate utilization, as well as the methyl-citrate pathway. Our study reveals that ethanolamine utilization bestows a competitive advantage of AIEC strains that are metabolically capable of its degradation in the presence of bile salts. We observed that bile salts activated secondary metabolism pathways that communicate to provide an energy benefit to AIEC. Bile salts may be used by AIEC as an environmental signal to promote their colonization.


Asunto(s)
Ácidos y Sales Biliares/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Acetilcoenzima A/metabolismo , Adaptación Fisiológica , Animales , Adhesión Bacteriana , Enfermedad de Crohn/microbiología , Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Etanolamina/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Genes Bacterianos/efectos de los fármacos , Interacciones Microbiota-Huesped/efectos de los fármacos , Humanos , Íleon/microbiología , Mucosa Intestinal/microbiología , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/genética , Ratones , Ratones Endogámicos C57BL , Propilenglicol/metabolismo , Regulación hacia Arriba/efectos de los fármacos
5.
Cells ; 8(1)2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30634511

RESUMEN

The intestinal mucosa of Crohn's disease (CD) patients is abnormally colonized with adherent-invasive Escherichia coli (AIEC) that are able to adhere to and to invade intestinal epithelial cells (IECs), to survive in macrophages, and to induce a pro-inflammatory response. AIEC persist in the intestine, and induce inflammation in CEABAC10 transgenic mice expressing human CAECAM6, the receptor for AIEC. SUMOylation is a eukaryotic-reversible post-translational modification, in which SUMO, an ubiquitin-like polypeptide, is covalently linked to target proteins. Here, we investigated the role of SUMOylation in host responses to AIEC infection. We found that infection with the AIEC LF82 reference strain markedly decreased the levels of SUMO-conjugated proteins in human intestinal epithelial T84 cells. This was also observed in IECs from LF82-infected CEABAC10 transgenic mice. LF82-induced deSUMOylation in IECs was due in part to increased level of microRNA (miR)-18, which targets PIAS3 mRNA encoding a protein involved in SUMOylation. Over-expression of SUMOs in T84 cells induced autophagy, leading to a significant decrease in the number of intracellular LF82. Consistently, a decreased expression of UBC9, a protein necessary for SUMOylation, was accompanied with a decrease of LF82-induced autophagy, increasing bacterial intracellular proliferation and inflammation. Finally, the inhibition of miR-18 significantly decreased the number of intracellular LF82. In conclusion, our results suggest that AIEC inhibits the autophagy response to replicate intracellularly by manipulating host SUMOylation.


Asunto(s)
Autofagia , Enfermedad de Crohn/microbiología , Infecciones por Escherichia coli/complicaciones , Mucosa Intestinal/metabolismo , Sumoilación , Animales , Adhesión Bacteriana , Línea Celular Tumoral , Enfermedad de Crohn/metabolismo , Escherichia coli/patogenicidad , Humanos , Ratones , Ratones Transgénicos , Proteínas Inhibidoras de STAT Activados/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo
6.
Toxins (Basel) ; 10(4)2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29642622

RESUMEN

Cyclomodulins are bacterial toxins that interfere with the eukaryotic cell cycle. A new cyclomodulin called colibactin, which is synthetized by the pks genomic island, was discovered in 2006. Despite many efforts, colibactin has not yet been purified, and its structure remains elusive. Interestingly, the pks island is found in members of the family Enterobacteriaceae (mainly Escherichia coli and Klebsiella pneumoniae) isolated from different origins, including from intestinal microbiota, septicaemia, newborn meningitis, and urinary tract infections. Colibactin-producing bacteria induce chromosomal instability and DNA damage in eukaryotic cells, which leads to senescence of epithelial cells and apoptosis of immune cells. The pks island is mainly observed in B2 phylogroup E. coli strains, which include extra-intestinal pathogenic E. coli strains, and pksE. coli are over-represented in biopsies isolated from colorectal cancer. In addition, pksE. coli bacteria increase the number of tumours in diverse colorectal cancer mouse models. Thus, colibactin could have a major impact on human health. In the present review, we will focus on the biological effects of colibactin, the distribution of the pks island, and summarize what is currently known about its synthesis and its structure.


Asunto(s)
Toxinas Bacterianas , Mutágenos , Péptidos , Policétidos , Animales , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidad , Islas Genómicas , Humanos , Mutágenos/química , Mutágenos/metabolismo , Mutágenos/toxicidad , Péptidos/química , Péptidos/metabolismo , Péptidos/toxicidad , Policétidos/química , Policétidos/metabolismo , Policétidos/toxicidad
7.
Toxins (Basel) ; 8(7)2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27429000

RESUMEN

Cytolethal distending toxin (CDT) is found in Gram-negative bacteria, especially in certain Proteobacteria such as the Pasteurellaceae family, including Haemophilus ducreyi and Aggregatibacter (Actinobacillus) actinomycetemcomitans, in the Enterobacteriaceae family and the Campylobacterales order, including the Campylobacter and Helicobacter species. In vitro and in vivo studies have clearly shown that this toxin has a strong effect on cellular physiology (inflammation, immune response modulation, tissue damage). Some works even suggest a potential involvement of CDT in cancers. In this review, we will discuss these different aspects.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Bacterias Gramnegativas/patogenicidad , Infecciones por Bacterias Gramnegativas/microbiología , Animales , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Daño del ADN , Bacterias Gramnegativas/inmunología , Bacterias Gramnegativas/metabolismo , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/metabolismo , Infecciones por Bacterias Gramnegativas/patología , Interacciones Huésped-Patógeno , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Neoplasias/metabolismo , Neoplasias/microbiología , Neoplasias/patología , Factores de Riesgo
8.
Gut ; 65(2): 278-85, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25588406

RESUMEN

OBJECTIVE: Colorectal cancers (CRCs) are frequently colonised by colibactin toxin-producing Escherichia coli bacteria that induce DNA damage in host cells and exhibit protumoural activities. Our objective was to identify small molecules inhibiting the toxic effects induced by these colibactin-producing bacteria. DESIGN: A structural approach was adopted for the identification of a putative ligand for the ClbP enzyme involved in the synthesis of colibactin. Intestinal epithelial cells and a CRC mouse model were used to assess the activity of the selected compounds in vitro and in vivo. RESULTS: Docking experiments identified two boron-based compounds with computed ligand efficiency values (-0.8 and -0.9 kcal/mol/atom) consistent with data expected for medicinal chemistry leads. The crystalline structure of ClbP in complex with the compounds confirmed that the compounds were binding to the active site of ClbP. The two compounds (2 mM) suppressed the genotoxic activity of colibactin-producing E coli both in vitro and in vivo. The mean degree of suppression of DNA damage for the most efficient compound was 98±2% (95% CI). This compound also prevented cell proliferation and colibactin-producing E coli-induced tumourigenesis in mice. In a CRC murine model colonised by colibactin-producing E coli, the number of tumours decreased by 3.5-fold in animals receiving the compound in drinking water (p<0.01). CONCLUSIONS: These results demonstrate that targeting colibactin production controls the genotoxic and protumoural effects induced by this toxin.


Asunto(s)
Ácidos Borónicos/farmacología , Neoplasias Colorrectales/prevención & control , Escherichia coli/efectos de los fármacos , Péptidos/metabolismo , Policétidos/metabolismo , Animales , Proteínas de Unión al Calcio/farmacología , Neoplasias Colorrectales/microbiología , Daño del ADN/fisiología , Escherichia coli/metabolismo , Ligandos , Ratones , Ratones Endogámicos BALB C , Mutágenos
9.
Cell Microbiol ; 18(5): 617-31, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26499863

RESUMEN

The aetiology of Crohn's disease (CD) involves disorders in host genetic factors and intestinal microbiota. Adherent-invasive Escherichia coli (AIEC) are receiving increased attention because in studies of mucosa-associated microbiota, they are more prevalent in CD patients than in healthy subjects. AIEC are associated both with ileal and colonic disease phenotypes. In this study, we reported a protease called Vat-AIEC from AIEC that favours the mucosa colonization. The deletion of the Vat-AIEC-encoding gene resulted in an adhesion-impaired phenotype in vitro and affected the colonization of bacteria in contact with intestinal epithelial cells in a murine intestinal loop model, and also their gut colonization in vivo. Furthermore, unlike LF82Δvat-AIEC, wild-type AIEC reference strain LF82 was able to penetrate a mucus column extensively and promoted the degradation of mucins and a decrease in mucus viscosity. Vat-AIEC transcription was stimulated by several chemical conditions found in the ileum environment. Finally, the screening of E. coli strains isolated from CD patients revealed a preferential vat-AIEC association with AIEC strains belonging to the B2 phylogroup. Overall, this study revealed a new component of AIEC virulence that might favour their implantation in the gut of CD patients.


Asunto(s)
Toxinas Bacterianas/genética , Enfermedad de Crohn/genética , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Microbioma Gastrointestinal/genética , Animales , Adhesión Bacteriana/genética , Toxinas Bacterianas/metabolismo , Enfermedad de Crohn/microbiología , Enfermedad de Crohn/patología , Células Epiteliales/microbiología , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Proteínas de Escherichia coli/metabolismo , Humanos , Íleon/microbiología , Íleon/patología , Mucosa Intestinal/microbiología , Ratones , Moco/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA