Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
World J Gastroenterol ; 30(21): 2817-2826, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38899326

RESUMO

BACKGROUND: The association between the intestinal microbiota and psychiatric disorders is becoming increasingly apparent. The gut microbiota contributes to colorectal carcinogenesis (CRC), as demonstrated with colibactin-producing Escherichia coli (CoPEC). AIM: To evaluate the association between CoPEC prevalence and anxiety- and depressive-like behaviors with both preclinical and clinical approaches. METHODS: Patients followed after a CRC surgery and for whom the prevalence of CoPEC has been investigated underwent a psychiatric interview. Results were compared according to the CoPEC colonization. In parallel C57BL6/J wild type mice and mice with a CRC susceptibility were chronically infected with a CoPEC strain. Their behavior was assessed using the Elevated Plus Maze test, the Forced Swimming Test and the Behavior recognition system PhenoTyper®. RESULTS: In a limited cohort, all patients with CoPEC colonization presented with psychiatric disorders several years before cancer diagnosis, whereas only one patient (17%) without CoPEC did. This result was confirmed in C57BL6/J wild-type mice and in a CRC susceptibility mouse model (adenomatous polyposis colimultiple intestinal neoplasia/+). Mice exhibited a significant increase in anxiety- and depressive-like behaviors after chronic infection with a CoPEC strain. CONCLUSION: This finding provides the first evidence that CoPEC infection can induce microbiota-gut-brain axis disturbances in addition to its procarcinogenic properties.


Assuntos
Ansiedade , Depressão , Modelos Animais de Doenças , Infecções por Escherichia coli , Microbioma Gastrointestinal , Camundongos Endogâmicos C57BL , Peptídeos , Policetídeos , Animais , Humanos , Masculino , Policetídeos/metabolismo , Depressão/psicologia , Depressão/microbiologia , Ansiedade/psicologia , Ansiedade/microbiologia , Ansiedade/etiologia , Camundongos , Feminino , Idoso , Pessoa de Meia-Idade , Infecções por Escherichia coli/psicologia , Infecções por Escherichia coli/microbiologia , Peptídeos/metabolismo , Escherichia coli/isolamento & purificação , Neoplasias do Colo/psicologia , Neoplasias do Colo/microbiologia , Prevalência , Eixo Encéfalo-Intestino
2.
Gut Microbes ; 16(1): 2320291, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38417029

RESUMO

Intratumoral bacteria flexibly contribute to cellular and molecular tumor heterogeneity for supporting cancer recurrence through poorly understood mechanisms. Using spatial metabolomic profiling technologies and 16SrRNA sequencing, we herein report that right-sided colorectal tumors are predominantly populated with Colibactin-producing Escherichia coli (CoPEC) that are locally establishing a high-glycerophospholipid microenvironment with lowered immunogenicity. It coincided with a reduced infiltration of CD8+ T lymphocytes that produce the cytotoxic cytokines IFN-γ where invading bacteria have been geolocated. Mechanistically, the accumulation of lipid droplets in infected cancer cells relied on the production of colibactin as a measure to limit genotoxic stress to some extent. Such heightened phosphatidylcholine remodeling by the enzyme of the Land's cycle supplied CoPEC-infected cancer cells with sufficient energy for sustaining cell survival in response to chemotherapies. This accords with the lowered overall survival of colorectal patients at stage III-IV who were colonized by CoPEC when compared to patients at stage I-II. Accordingly, the sensitivity of CoPEC-infected cancer cells to chemotherapies was restored upon treatment with an acyl-CoA synthetase inhibitor. By contrast, such metabolic dysregulation leading to chemoresistance was not observed in human colon cancer cells that were infected with the mutant strain that did not produce colibactin (11G5∆ClbQ). This work revealed that CoPEC locally supports an energy trade-off lipid overload within tumors for lowering tumor immunogenicity. This may pave the way for improving chemoresistance and subsequently outcome of CRC patients who are colonized by CoPEC.


Assuntos
Neoplasias Colorretais , Microbioma Gastrointestinal , Peptídeos , Policetídeos , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Microambiente Tumoral , Resistencia a Medicamentos Antineoplásicos , Mutagênicos/metabolismo , Recidiva Local de Neoplasia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/microbiologia , Policetídeos/metabolismo , Lipídeos
3.
Gut Microbes ; 16(1): 2310215, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38374654

RESUMO

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.


Assuntos
Microbioma Gastrointestinal , Neoplasias , Peptídeos , Policetídeos , Humanos , Camundongos , Animais , Escherichia coli/genética , Escherichia coli/metabolismo , Transição Epitelial-Mesenquimal , Mutagênicos/metabolismo , Policetídeos/farmacologia , Policetídeos/metabolismo , Modelos Animais de Doenças , Células-Tronco Neoplásicas/metabolismo
4.
Gut Microbes ; 15(1): 2229569, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37417545

RESUMO

Colorectal cancer (CRC) patients are frequently colonized by colibactin-producing Escherichia coli (CoPEC) (>40%), which enhances tumorigenesis in mouse models of CRC. We observed that 50% of CoPEC also contains the cnf1 gene, which encodes cytotoxic necrotizing factor-1 (CNF1), an enhancer of the eukaryotic cell cycle. The impact of its co-occurrence with colibactin (Clb) has not yet been investigated. We evaluated the impact of CNF1 on colorectal tumorigenesis using human colonic epithelial HT-29 cells and CRC-susceptible ApcMin/+ mice inoculated with the CoPEC 21F8 clinical strain (Clb+Cnf+) or 21F8 isogenic mutants (Clb+Cnf-, Clb-Cnf+ and Clb-Cnf-). Infection with the Clb+Cnf- strain induced higher levels of inflammatory cytokines and senescence markers both in vitro and in vivo compared to those induced by infection with the Clb+Cnf+ strain. In contrast, the Clb+Cnf- and Clb+Cnf+ strains generated similar levels of DNA damage in HT-29 cells and in colonic murine tissues. Furthermore, the ApcMin/+ mice inoculated with the Clb+Cnf- strain developed significantly more tumors than the mice inoculated with the Clb+Cnf+ strain or the isogenic mutants, and the composition of their microbiota was changed. Finally, rectal administration of the CNF1 protein in ApcMin/+ mice inoculated with the Clb+Cnf- strain significantly decreased tumorigenesis and inflammation. Overall, this study provides evidence that CNF1 decreases the carcinogenic effects of CoPEC in ApcMin/+ mice by decreasing CoPEC-induced cellular senescence and inflammation.


Assuntos
Infecções por Escherichia coli , Proteínas de Escherichia coli , Microbioma Gastrointestinal , Camundongos , Humanos , Animais , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Colo , Carcinogênese , Transformação Celular Neoplásica , Inflamação
5.
Int J Mol Sci ; 22(7)2021 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-33805299

RESUMO

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.


Assuntos
Autofagia , Doença de Crohn/etiologia , Infecções por Escherichia coli/complicações , Escherichia coli/patogenicidade , Mucosa Intestinal/fisiopatologia , Fenóis/metabolismo , Tiazóis/metabolismo , Animais , Doença de Crohn/fisiopatologia , Escherichia coli/metabolismo , Infecções por Escherichia coli/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos
6.
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
7.
Syst Appl Microbiol ; 43(5): 126124, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32847790

RESUMO

Polyphasic taxonomic analysis was performed on a novel bacterium, designated UR159T, isolated in 2016 from human blood of a septic patient hospitalized in France. Preliminary 16S rRNA gene sequence-based phylogenetic analysis indicated that strain UR159T belonged to the family Flavobacteriaceae, forming a distinct phyletic line distantly related (<94% sequence similarity) to known species of the family. Further phenotypic, chemotaxonomic and genomic analyses were performed. Cells were non-motile, oxidase-negative, catalase-positive Gram-negative rods. It was strictly aerobic yielding yellow-pigmented colonies, and was metabolically rather inert. Major fatty acids were iso-branched fatty acids, predominantly iso-C15:0 (55.5%) and iso-C17:1ω9c (8.8%). Whole genome sequencing revealed a 2.3-Mbp genome encoding a total of 2262 putative genes with a genomic DNA G+C content at 37.6mol%. The average nucleotide identity (ANI) and in silico DNA-DNA hybridization (isDDH) values between strain UR159T and the most closely related members of the Flavobacteriaceae family were <75% and <39%, respectively, much below the established cut-offs for ANI (<95-96%) and isDDH (<70%) for species and genus delineation. Average Amino Acid Identity (AAI) percentages were also estimated and were lower than 65% (cut-off proposed for genus delineation for uncultivated prokaryotes) in all cases, except for F. marinum that was just at the limit (65.1%). Based on these findings, we propose it as a new genus and species, Avrilella dinanensis gen. nov., sp. nov. (type strain UR159T=CIP 111616T=DSM 105483T).


Assuntos
Sangue/microbiologia , Flavobacteriaceae/classificação , Flavobacteriaceae/isolamento & purificação , Sepse/microbiologia , Aerobiose , Idoso de 80 Anos ou mais , Aminoácidos/análise , Composição de Bases , DNA Bacteriano/química , DNA Bacteriano/genética , Ácidos Graxos/análise , Feminino , Flavobacteriaceae/genética , Flavobacteriaceae/fisiologia , Genes Bacterianos , Genes de RNAr , Genoma Bacteriano , Genômica , Humanos , Fenótipo , Filogenia , Pigmentação , RNA Ribossômico 16S/genética , Sequenciamento Completo do Genoma
8.
Nature ; 580(7802): 269-273, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32106218

RESUMO

Various species of the intestinal microbiota have been associated with the development of colorectal cancer1,2, but it has not been demonstrated that bacteria have a direct role in the occurrence of oncogenic mutations. Escherichia coli can carry the pathogenicity island pks, which encodes a set of enzymes that synthesize colibactin3. This compound is believed to alkylate DNA on adenine residues4,5 and induces double-strand breaks in cultured cells3. Here we expose human intestinal organoids to genotoxic pks+ E. coli by repeated luminal injection over five months. Whole-genome sequencing of clonal organoids before and after this exposure revealed a distinct mutational signature that was absent from organoids injected with isogenic pks-mutant bacteria. The same mutational signature was detected in a subset of 5,876 human cancer genomes from two independent cohorts, predominantly in colorectal cancer. Our study describes a distinct mutational signature in colorectal cancer and implies that the underlying mutational process results directly from past exposure to bacteria carrying the colibactin-producing pks pathogenicity island.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/microbiologia , Escherichia coli/genética , Escherichia coli/patogenicidade , Ilhas Genômicas/genética , Mutagênese , Mutação , Técnicas de Cocultura , Estudos de Coortes , Sequência Consenso , Dano ao DNA , Microbioma Gastrointestinal , Humanos , Organoides/citologia , Organoides/metabolismo , Organoides/microbiologia , Peptídeos/genética , Policetídeos
9.
Gastroenterology ; 158(5): 1373-1388, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31917256

RESUMO

BACKGROUND & AIMS: Colibactin-producing Escherichia coli (CoPEC) colonize the colonic mucosa of a higher proportion of patients with vs without colorectal cancer (CRC) and promote colorectal carcinogenesis in susceptible mouse models of CRC. Autophagy degrades cytoplasmic contents, including intracellular pathogens, via lysosomes and regulates intestinal homeostasis. We investigated whether inhibiting autophagy affects colorectal carcinogenesis in susceptible mice infected with CoPEC. METHODS: Human intestinal epithelial cells (IECs) (HCT-116) were infected with a strain of CoPEC (11G5 strain) isolated from a patient or a mutant strain that does not produce colibactin (11G5ΔclbQ). Levels of ATG5, ATG16L1, and SQSTM1 (also called p62) were knocked down in HCT-116 cells using small interfering RNAs. ApcMin/+ mice and ApcMin/+ mice with IEC-specific disruption of Atg16l1 (ApcMin/+/Atg16l1ΔIEC) were infected with 11G5 or 11G5ΔclbQ. Colonic tissues were collected from mice and analyzed for tumor size and number and by immunohistochemical staining, immunoblot, and quantitative reverse transcription polymerase chain reaction for markers of autophagy, DNA damage, cell proliferation, and inflammation. We analyzed levels of messenger RNAs (mRNAs) encoding proteins involved in autophagy in colonic mucosal tissues from patients with sporadic CRC colonized with vs without CoPEC by quantitative reverse-transcription polymerase chain reaction. RESULTS: Patient colonic mucosa with CoPEC colonization had higher levels of mRNAs encoding proteins involved in autophagy than colonic mucosa without these bacteria. Infection of cultured IECs with 11G5 induced autophagy and DNA damage repair, whereas infection with 11G5ΔclbQ did not. Knockdown of ATG5 in HCT-116 cells increased numbers of intracellular 11G5, secretion of interleukin (IL) 6 and IL8, and markers of DNA double-strand breaks but reduced markers of DNA repair, indicating that autophagy is required for bacteria-induced DNA damage repair. Knockdown of ATG5 in HCT-116 cells increased 11G5-induced senescence, promoting proliferation of uninfected cells. Under uninfected condition, ApcMin/+/Atg16l1ΔIEC mice developed fewer and smaller colon tumors than ApcMin/+ mice. However, after infection with 11G5, ApcMin/+/Atg16l1ΔIEC mice developed more and larger tumors, with a significant increase in mean histologic score, than infected ApcMin/+ mice. Increased levels of Il6, Tnf, and Cxcl1 mRNAs, decreased level of Il10 mRNA, and increased markers of DNA double-strand breaks and proliferation were observed in the colonic mucosa of 11G5-infected ApcMin/+/Atg16l1ΔIEC mice vs 11G5-infected ApcMin/+ mice. CONCLUSION: Infection of IECs and susceptible mice with CoPEC promotes autophagy, which is required to prevent colorectal tumorigenesis. Loss of ATG16L1 from IECs increases markers of inflammation, DNA damage, and cell proliferation and increases colorectal tumorigenesis in 11G5-infected ApcMin/+ mice. These findings indicate the importance of autophagy in response to CoPEC infection, and strategies to induce autophagy might be developed for patients with CRC and CoPEC colonization.


Assuntos
Autofagia , Carcinogênese/imunologia , Colo/microbiologia , Neoplasias do Colo/imunologia , Mucosa Intestinal/microbiologia , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Proteínas Relacionadas à Autofagia/genética , Proteínas Relacionadas à Autofagia/imunologia , Proteínas Relacionadas à Autofagia/metabolismo , Carcinogênese/efeitos dos fármacos , Proliferação de Células , Colo/imunologia , Colo/patologia , Neoplasias do Colo/genética , Neoplasias do Colo/microbiologia , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/patologia , Escherichia coli/imunologia , Escherichia coli/isolamento & purificação , Escherichia coli/patogenicidade , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HCT116 , Células HeLa , Interações Hospedeiro-Patógeno/imunologia , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Transgênicos , Peptídeos/toxicidade , Policetídeos/toxicidade , RNA Interferente Pequeno/metabolismo
10.
Sci Rep ; 9(1): 2175, 2019 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-30778122

RESUMO

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.


Assuntos
Ácidos e Sais Biliares/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Acetilcoenzima A/metabolismo , Adaptação Fisiológica , Animais , Aderência Bacteriana , Doença de Crohn/microbiologia , Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Etanolamina/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Genes Bacterianos/efeitos dos fármacos , Interações entre Hospedeiro e Microrganismos/efeitos dos fármacos , Humanos , Íleo/microbiologia , Mucosa Intestinal/microbiologia , Redes e Vias Metabólicas/efeitos dos fármacos , Redes e Vias Metabólicas/genética , Camundongos , Camundongos Endogâmicos C57BL , Propilenoglicol/metabolismo , Regulação para Cima/efeitos dos fármacos
11.
Cells ; 8(1)2019 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-30634469

RESUMO

Crohn's disease is characterized by abnormal ileal colonization by adherent-invasive E. coli (AIEC) and expansion of mesenteric adipose tissue. This study assessed the preventive effect of spontaneous physical activity (PA) on the gut-adipose tissue in a mouse model that mimics Crohn's disease susceptibility. Thirty-five CEABAC10 male mice performed spontaneous PA (wheel group; n = 24) or not (controls; n = 11) for 12 weeks. At week 12, mice were orally challenged with the AIEC LF82 strain for 6 days. Body composition, glycaemic control, intestinal permeability, gut microbiota composition, and fecal short-chain fatty acids were assessed in both groups. Animals were fed a high fat/high sugar diet throughout the study. After exposure to AIEC, mesenteric adipose tissue weight was lower in the wheel group. Tight junction proteins expression increased with spontaneous PA, whereas systemic lipopolysaccharides were negatively correlated with the covered distance. Bifidobacterium and Lactobacillus decreased in controls, whereas Oscillospira and Ruminococcus increased in the wheel group. Fecal propionate and butyrate were also higher in the wheel group. In conclusion, spontaneous physical activity promotes healthy gut microbiota composition changes and increases short-chain fatty acids in CEABAC10 mice fed a Western diet and exposed to AIEC to mimic Crohn's disease.


Assuntos
Tecido Adiposo/metabolismo , Doença de Crohn/prevenção & controle , Intestinos/microbiologia , Esforço Físico , Animais , Aderência Bacteriana , Doença de Crohn/microbiologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Escherichia coli/patogenicidade , Infecções por Escherichia coli/metabolismo , Ácidos Graxos Voláteis/metabolismo , Feminino , Microbioma Gastrointestinal , Glucose/metabolismo , Masculino , Camundongos , Camundongos Transgênicos
12.
Cells ; 8(1)2019 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-30634511

RESUMO

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.


Assuntos
Autofagia , Doença de Crohn/microbiologia , Infecções por Escherichia coli/complicações , Mucosa Intestinal/metabolismo , Sumoilação , Animais , Aderência Bacteriana , Linhagem Celular Tumoral , Doença de Crohn/metabolismo , Escherichia coli/patogenicidade , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Inibidoras de STAT Ativados/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo
13.
Toxins (Basel) ; 10(4)2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29642622

RESUMO

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.


Assuntos
Toxinas Bacterianas , Mutagênicos , Peptídeos , Policetídeos , Animais , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidade , Ilhas Genômicas , Humanos , Mutagênicos/química , Mutagênicos/metabolismo , Mutagênicos/toxicidade , Peptídeos/química , Peptídeos/metabolismo , Peptídeos/toxicidade , Policetídeos/química , Policetídeos/metabolismo , Policetídeos/toxicidade
14.
Dig Dis Sci ; 63(4): 870-880, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29357083

RESUMO

BACKGROUND: Niemann-Pick disease, type C (NPC) is a rare lysosomal storage disorder characterized by progressive neurodegeneration, splenomegaly, hepatomegaly, and early death. NPC is caused by mutations in either the NPC1 or NPC2 gene. Impaired NPC function leads to defective intracellular transport of unesterified cholesterol and its accumulation in late endosomes and lysosomes. A high frequency of Crohn disease has been reported in NPC1 patients, suggesting that gastrointestinal tract pathology may become a more prominent clinical issue if effective therapies are developed to slow the neurodegeneration. The Npc1 nih mouse model on a BALB/c background replicates the hepatic and neurological disease observed in NPC1 patients. Thus, we sought to characterize the gastrointestinal tract pathology in this model to determine whether it can serve as a model of Crohn disease in NPC1. METHODS: We analyzed the gastrointestinal tract and isolated macrophages of BALB/cJ cNctr-Npc1m1N/J (Npc1-/-) mouse model to determine whether there was any Crohn-like pathology or inflammatory cell activation. We also evaluated temporal changes in the microbiota by 16S rRNA sequencing of fecal samples to determine whether there were changes consistent with Crohn disease. RESULTS: Relative to controls, Npc1 mutant mice demonstrate increased inflammation and crypt abscesses in the gastrointestinal tract; however, the observed pathological changes are significantly less than those observed in other Crohn disease mouse models. Analysis of Npc1 mutant macrophages demonstrated an increased response to lipopolysaccharides and delayed bactericidal activity; both of which are pathological features of Crohn disease. Analysis of the bacterial microbiota does not mimic what is reported in Crohn disease in either human or mouse models. We did observe significant increases in cyanobacteria and epsilon-proteobacteria. The increase in epsilon-proteobacteria may be related to altered cholesterol homeostasis since cholesterol is known to promote growth of this bacterial subgroup. CONCLUSIONS: Macrophage dysfunction in the BALB/c Npc1-/- mouse is similar to that observed in other Crohn disease models. However, neither the degree of pathology nor the microbiota changes are typical of Crohn disease. Thus, this mouse model is not a good model system for Crohn disease pathology reported in NPC1 patients.


Assuntos
Doença de Crohn/etiologia , Doença de Crohn/patologia , Trato Gastrointestinal/patologia , Doença de Niemann-Pick Tipo C/patologia , Animais , Modelos Animais de Doenças , Trato Gastrointestinal/microbiologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos BALB C , Doença de Niemann-Pick Tipo C/microbiologia
16.
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
17.
J Antimicrob Chemother ; 72(2): 402-406, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27793962

RESUMO

OBJECTIVES: To investigate the resistance mechanisms and genetic support underlying the high resistance level of the Klebsiella pneumoniae strain CMUL78 to aminoglycoside and ß-lactam antibiotics. METHODS: Antibiotic susceptibility was assessed by the disc diffusion method and MICs were determined by the microdilution method. Antibiotic resistance genes and their genetic environment were characterized by PCR and Sanger sequencing. Plasmid contents were analysed in the clinical strain and transconjugants obtained by mating-out assays. Complete plasmid sequencing was performed with PacBio and Illumina technology. RESULTS: Strain CMUL78 co-produced the 16S rRNA methyltransferase (RMTase) RmtH, carbapenemase OXA-48 and ESBL SHV-12. The rmtH- and blaSHV-12-encoding genes were harboured by a novel ∼115 kb IncFIIk plasmid designated pRmtH, and blaOXA-48 by a ∼62 kb IncL/M plasmid related to pOXA-48a. pRmtH plasmid possessed seven different stability modules, one of which is a novel hybrid toxin-antitoxin system. Interestingly, pRmtH plasmid harboured a 4-fold amplification of an rmtH-ISCR2 unit arranged in tandem and inserted within a novel IS26-based composite transposon designated Tn6329. CONCLUSIONS: This is the first known report of the 16S RMTase-encoding gene rmtH in a plasmid. The rmtH-ISCR2 unit was inserted in a composite transposon as a 4-fold tandem repeat, a scarcely reported organization.


Assuntos
Aminoglicosídeos/farmacologia , Farmacorresistência Bacteriana , Sequências Repetitivas Dispersas , Klebsiella pneumoniae/efeitos dos fármacos , Metiltransferases/genética , Plasmídeos/análise , beta-Lactamas/farmacologia , Conjugação Genética , DNA Bacteriano/química , DNA Bacteriano/genética , Transferência Genética Horizontal , Humanos , Recém-Nascido , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/isolamento & purificação , Testes de Sensibilidade Microbiana , Plasmídeos/classificação , Reação em Cadeia da Polimerase , Análise de Sequência de DNA
18.
Toxins (Basel) ; 8(7)2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27429000

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Bactérias Gram-Negativas/patogenicidade , Infecções por Bactérias Gram-Negativas/microbiologia , Animais , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Dano ao DNA , Bactérias Gram-Negativas/imunologia , Bactérias Gram-Negativas/metabolismo , Infecções por Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/metabolismo , Infecções por Bactérias Gram-Negativas/patologia , Interações Hospedeiro-Patógeno , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Neoplasias/metabolismo , Neoplasias/microbiologia , Neoplasias/patologia , Fatores de Risco
19.
Med Sci (Paris) ; 32(2): 175-82, 2016 Feb.
Artigo em Francês | MEDLINE | ID: mdl-26936175

RESUMO

The gut microbiota maintains a relationship with its host with strong mutual benefits. Changes in the composition of the intestinal microbiota have been detected in colorectal cancer patients to the extent that it is now considered as a real contributing factor in this pathology. In this review, we focus on three commensal bacterial species, namely Bacteroides fragilis, Fusobacterium nucleatum, and Escherichia coli, which seem to emerge as pathogens and to contribute to colorectal carcinogenesis through their inflammatory and oncogenic properties.


Assuntos
Fenômenos Fisiológicos Bacterianos , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/patologia , Simbiose/fisiologia , Animais , Bacteroides fragilis/patogenicidade , Carcinogênese/patologia , Progressão da Doença , Escherichia coli/patogenicidade , Fusobacterium nucleatum/patogenicidade , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Humanos
20.
World J Gastroenterol ; 22(2): 501-18, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26811603

RESUMO

The gut microbiota acts as a real organ. The symbiotic interactions between resident micro-organisms and the digestive tract highly contribute to maintain the gut homeostasis. However, alterations to the microbiome caused by environmental changes (e.g., infection, diet and/or lifestyle) can disturb this symbiotic relationship and promote disease, such as inflammatory bowel diseases and cancer. Colorectal cancer is a complex association of tumoral cells, non-neoplastic cells and a large amount of micro-organisms, and the involvement of the microbiota in colorectal carcinogenesis is becoming increasingly clear. Indeed, many changes in the bacterial composition of the gut microbiota have been reported in colorectal cancer, suggesting a major role of dysbiosis in colorectal carcinogenesis. Some bacterial species have been identified and suspected to play a role in colorectal carcinogenesis, such as Streptococcus bovis, Helicobacter pylori, Bacteroides fragilis, Enterococcus faecalis, Clostridium septicum, Fusobacterium spp. and Escherichia coli. The potential pro-carcinogenic effects of these bacteria are now better understood. In this review, we discuss the possible links between the bacterial microbiota and colorectal carcinogenesis, focusing on dysbiosis and the potential pro-carcinogenic properties of bacteria, such as genotoxicity and other virulence factors, inflammation, host defenses modulation, bacterial-derived metabolism, oxidative stress and anti-oxidative defenses modulation. We lastly describe how bacterial microbiota modifications could represent novel prognosis markers and/or targets for innovative therapeutic strategies.


Assuntos
Bactérias/patogenicidade , Neoplasias Colorretais/microbiologia , Microbioma Gastrointestinal , Intestinos/microbiologia , Animais , Bactérias/metabolismo , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Dano ao DNA , Disbiose , Interações Hospedeiro-Patógeno , Humanos , Mediadores da Inflamação/metabolismo , Estresse Oxidativo , Fatores de Virulência/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA