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











Base de dados
Intervalo de ano de publicação
1.
J Nat Prod ; 83(12): 3614-3622, 2020 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-33270444

RESUMO

Lythrum salicaria herb (LSH) was applied in diarrhea therapy since ancient times. Despite empirically referenced therapeutic effects, the bioactivity mechanisms and chemical constituents responsible for pharmacological activity remain not fully resolved. Taking into consideration the historical use of LSH in treatment of diarrhea in humans and farm animals, the aim of the study was to examine in vitro the influence of LSH and its C-glycosylic ellagitannins on processes associated with maintaining intestinal epithelium integrity and enteropathogenic Escherichia coli (EPEC) growth and adhesion. LSH was not only inhibiting EPEC growth in a concentration dependent manner but also its adhesion to IPEC-J2 intestinal epithelial cell monolayers. Inhibitory activity toward EPEC growth was additionally confirmed ex vivo in distal colon samples of postweaning piglets. LSH and its dominating C-glycosylic ellagitannins, castalagin (1), vescalagin (2), and salicarinins A (3) and B (4) were stimulating IPEC-J2 monolayer formation by enhancing claudin 4 production. Parallelly tested gut microbiota metabolites of LSH ellagitannins, urolithin C (5), urolithin A (6), and its glucuronides (7) were inactive. The activities of LSH and the isolated ellagitannins support its purported antidiarrheal properties and indicate potential mechanisms responsible for its beneficial influence on the intestinal epithelium.


Assuntos
Aderência Bacteriana/efeitos dos fármacos , Escherichia coli Enteropatogênica/efeitos dos fármacos , Taninos Hidrolisáveis/farmacologia , Lythrum/química , Linhagem Celular , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Escherichia coli Enteropatogênica/fisiologia , Células Epiteliais/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos
2.
Vet Microbiol ; 241: 108555, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31928702

RESUMO

Avian pathogenic Escherichia coli (APEC) causes avian colibacillosis in poultry, which is characterized by systemic infections such as septicemia, air sacculitis, and pericarditis. APEC uses two-component regulatory systems (TCSs) to handle the stressful environments present in infected hosts. While many TCSs in E. coli have been well characterized, the RstA/RstB system in APEC has not been thoroughly investigated. The involvement of the RstA regulator in APEC pathogenesis was demonstrated during previous studies investigating its role in APEC persistence in chicken macrophages and respiratory infections. However, the mechanism underlying this phenomenon has not been clarified. Transcriptional analysis of the effect of rstAB deletion was therefore performed to improve the understanding of the RstA/RstB regulatory mechanism, and particularly its role in virulence. The transcriptomes of the rstAB mutant and the wild-type strain E058 were compared during their growth in the bloodstreams of challenged chickens. Overall, 198 differentially expressed (DE) genes were identified, and these indicated that RstA/RstB mainly regulates systems involved in nitrogen metabolism, iron acquisition, and acid resistance. Phenotypic assays indicated that the rstAB mutant responded more to an acidic pH than the wild-type strain did, possibly because of the repression of the acid-resistance operons hdeABD and gadABE by the deletion of rstAB. Based on the reported RstA box motif TACATNTNGTTACA, we identified four possible RstA target genes (hdeD, fadE, narG, and metE) among the DE genes. An electrophoretic mobility shift assay confirmed that RstA binds directly to the promoter of hdeD, and ß-galactosidase assays showed that hdeD expression was reduced by rstAB deletion, indicating that RstA directly regulates hdeD expression. The hdeD mutation resulted in virulence attenuation in both cultured chicken macrophages and experimentally infected chickens. In conclusion, our data suggest that RstA affects APEC E058 virulence partly by directly regulating the acidic resistance gene hdeD.


Assuntos
Escherichia coli Enteropatogênica/patogenicidade , Proteínas de Escherichia coli/análise , Macrófagos/microbiologia , Proteínas de Membrana/fisiologia , Animais , Galinhas , Biologia Computacional , Meios de Cultura/química , Escherichia coli Enteropatogênica/genética , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/veterinária , Proteínas de Escherichia coli/fisiologia , Deleção de Genes , Expressão Gênica , Concentração de Íons de Hidrogênio , Análise em Microsséries/veterinária , Mutação , Nitrogênio/deficiência , Doenças das Aves Domésticas/microbiologia , RNA Bacteriano/química , RNA Bacteriano/isolamento & purificação , RNA Complementar/química , RNA Complementar/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Organismos Livres de Patógenos Específicos , Virulência , beta-Galactosidase/metabolismo
3.
Int J Mol Sci ; 20(22)2019 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-31726759

RESUMO

LF82, a prototype of adherent-invasive E. coli (AIEC), is able to adhere to, invade, survive and replicate into intestinal epithelial cells. LF82 is able to enhance either its adhesion and invasion by up-regulating carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM-6), the main cell surface molecule for bacterial adhesion, and its intracellular survival by inducing host DNA damage, thus blocking the cellular cycle. Lactoferrin (Lf) is a multifunctional cationic glycoprotein of natural immunity, exerting an anti-invasive activity against LF82 when added to Caco-2 cells at the moment of infection. Here, the infection of 12 h Lf pre-treated Caco-2 cells was carried out at a time of 0 or 3 or 10 h after Lf removal from culture medium. The effect of Lf pre-treatment on LF82 invasiveness, survival, cell DNA damage, CEACAM-6 expression, apoptosis induction, as well as on Lf subcellular localization, has been evaluated. Lf, even if removed from culture medium, reduced LF82 invasion and survival as well as bacteria-induced DNA damage in Caco-2 cells independently from induction of apoptosis, modulation of CEACAM-6 expression and Lf sub-cellular localization. At our knowledge, this is the first study showing that the sole Lf pre-treatment can activate protective intracellular pathways, reducing LF82 invasiveness, intracellular survival and cell-DNA damages.


Assuntos
Diferenciação Celular , Dano ao DNA , Enterócitos , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Infecções por Escherichia coli , Lactoferrina/farmacologia , Animais , Células CACO-2 , Bovinos , Enterócitos/metabolismo , Enterócitos/microbiologia , Enterócitos/patologia , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/patologia , Humanos
4.
J Pept Sci ; 25(3): e3149, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30746861

RESUMO

Human pathogenic gram-negative bacteria, such as enteropathogenic Escherichia coli (EPEC), rely on type III secretion systems (T3SS) to translocate virulence factors directly into host cells. The coiled-coil domains present in the structural proteins of T3SS are conformed by amphipathic alpha-helical structures that play an important role in the protein-protein interaction and are essential for the assembly of the translocation complex. To investigate the inhibitory capacity of these domains on the T3SS of EPEC, we synthesized peptides between 7 and 34 amino acids based on the coiled-coil domains of proteins that make up this secretion system. This analysis was performed through in vitro hemolysis assays by assessing the reduction of T3SS-dependent red blood cell lysis in the presence of the synthesized peptides. After confirming its inhibitory capacity, we performed molecular modeling assays using combined techniques, docking-molecular dynamic simulations, and quantum-mechanic calculations of the various peptide-protein complexes, to improve the affinity of the peptides to the target proteins selected from T3SS. These techniques allowed us to demonstrate that the peptides with greater inhibitory activity, directed against the coiled-coil domain of the C-terminal region of EspA, present favorable hydrophobic and hydrogen bond molecular interactions. Particularly, the hydrogen bond component is responsible for the stabilization of the peptide-protein complex. This study demonstrates that compounds targeting T3SS from pathogenic bacteria can indeed inhibit bacterial infection by presenting a higher specificity than broad-spectrum antibiotics. In turn, these peptides could be taken as initial structures to design and synthesize new compounds that mimic their inhibitory pharmacophoric pattern.


Assuntos
Antibacterianos/farmacologia , Escherichia coli Enteropatogênica/efeitos dos fármacos , Escherichia coli Enteropatogênica/metabolismo , Peptídeos/farmacologia , Sistemas de Secreção Tipo III/efeitos dos fármacos , Antibacterianos/síntese química , Antibacterianos/química , Dicroísmo Circular , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Humanos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Peptídeos/síntese química , Peptídeos/química , Termodinâmica
5.
Microb Pathog ; 128: 396-404, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30660737

RESUMO

BACKGROUND: Gastric fluid pH serves an important function as an ecological filter to kill unwanted microbial taxa that would otherwise colonise the intestines, thereby shaping the diversity and composition of microbial communities found in the gut. The typical American-based diet causes the gastric pH to increase to pH 4 to 5, and it takes ∼2 h to return to pH 1.5 (normal). This window of increased gastric pH may allow potential pathogens to negotiate the hostile environment of the stomach. Another factor to consider is that in developing countries many people experience hypochlorhydria related to malnutrition and various gastric diseases. Enteropathogenic E. coli (EPEC) is a leading cause of infantile diarrhoea and has a high incidence in the developing world. The aim of this study was to assess the survival and recovery of non-acid adapted EPEC exposed to simulated gastric fluid (SGF) over a period of 180 min. RESULTS: EPEC were grown in nutrient-rich medium and acid challenged in SGF at pH 1.5, 2.5, 3.5 and 4.5. Culturability was evaluated using a standard plate count method, and metabolic viability was assessed via cellular energy (adenosine triphosphate [ATP] assay) and respiratory activity (3-bis(2-methyloxy-4-nitro-5-sulfophenyl)-5-[(phenylamino)carbonyl]-2H-tetrazolium hydroxide [XTT] assay), and recovery and proliferation by means of optical density in liquid cultures. Sampling was performed at 0, 30, 60, 120, and 180 min post-SGF exposure. The results of this study showed that EPEC is remarkably acid resistant and was able to survive a simulated gastric environment for up to 3 h (180 min) at various pH (1.5, 2.5, 3.5, and 4.5). EPEC was culturable at all pH (1.5, 2.5, 3.5 and 4.5) at the higher inoculum size of 5.4-7.1 × 106 CFU/ml, and at all pH except pH 1.5 at the lower inoculums of 5.4-7.1 × 103 CFU/ml or 5.4-7.1 × 101 CFU/ml. The organism remained metabolically viable at pH 1.5, 2.5, 3.5, and 4.5 and was able to recover and proliferate once placed in a neutral, nutrient-rich environment. CONCLUSION: In this study, EPEC demonstrated remarkable acid resistance and recovery at low pH without prior acid adaptation, which could prove to be problematic even in healthy people. In individuals with decreased gastric acidity, there is a higher probability of pathogen colonization and a resulting change in the gut microbiome. The results highlight the potential increase of food- and waterborne diseases in persons with compromised gastric function, or who are malnourished or immunocompromised. The data herein may possibly help in calculating more precisely the risk associated with consuming bacterial contaminated food and water in these individuals.


Assuntos
Adaptação Fisiológica/fisiologia , Farmacorresistência Bacteriana/fisiologia , Escherichia coli Enteropatogênica/efeitos dos fármacos , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Escherichia coli Enteropatogênica/fisiologia , Ácido Gástrico , Viabilidade Microbiana/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Proliferação de Células/efeitos dos fármacos , Contagem de Colônia Microbiana , Infecções por Escherichia coli/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Estômago/microbiologia , Estresse Fisiológico , Fatores de Tempo
6.
PLoS One ; 11(6): e0157334, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27309855

RESUMO

Interference with bacterial quorum sensing communication provides an anti-virulence strategy to control pathogenic bacteria. Here, using the Enteropathogenic E. coli (EPEC) O103:H2, we showed for the first time that thiophenone TF101 reduced expression of lsrB; the gene encoding the AI-2 receptor. Combined results of transcriptional and phenotypic analyses suggested that TF101 interfere with AI-2 signalling, possibly by competing with AI-2 for binding to LsrB. This is supported by in silico docking prediction of thiophenone TF101 in the LsrB pocket. Transcriptional analyses furthermore showed that thiophenone TF101 interfered with expression of the virulence genes eae and fimH. In addition, TF101 reduced AI-2 induced E. coli adhesion to colorectal adenocarcinoma cells. TF101, on the other hand, did not affect epinephrine or norepinephrine enhanced E. coli adhesion. Overall, our results showed that thiophenone TF101 interfered with virulence expression in E. coli O103:H2, suggestedly by interfering with AI-2 mediated quorum sensing. We thus conclude that thiophenone TF101 might represent a promising future anti-virulence agent in the fight against pathogenic E. coli.


Assuntos
Antibacterianos/farmacologia , Escherichia coli Enteropatogênica/efeitos dos fármacos , Escherichia coli Enteropatogênica/patogenicidade , Regulação Bacteriana da Expressão Gênica , Homosserina/análogos & derivados , Lactonas/antagonistas & inibidores , Tiofenos/farmacologia , Adesinas Bacterianas/química , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Adesinas de Escherichia coli/química , Adesinas de Escherichia coli/genética , Adesinas de Escherichia coli/metabolismo , Antibacterianos/química , Aderência Bacteriana/efeitos dos fármacos , Sítios de Ligação , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Escherichia coli Enteropatogênica/genética , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Epinefrina/farmacologia , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Fímbrias/química , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/metabolismo , Homosserina/antagonistas & inibidores , Homosserina/metabolismo , Humanos , Lactonas/metabolismo , Simulação de Acoplamento Molecular , Norepinefrina/farmacologia , Ligação Proteica , Percepção de Quorum/efeitos dos fármacos , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Tiofenos/química , Virulência
7.
Infect Immun ; 83(1): 379-88, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25385791

RESUMO

Enteropathogenic Escherichia coli (EPEC) is classified as typical (tEPEC) or atypical (aEPEC) based on the presence or absence of the E. coli adherence factor plasmid (pEAF), respectively. The hallmark of EPEC infection is the formation of the attaching and effacing (A/E) lesions on the gut mucosa. We compared the kinetics of A/E lesion formation induced by aEPEC and tEPEC. The examination of infected HEp-2 cells clearly demonstrated delayed A/E lesion formation by aEPEC in comparison to tEPEC. This delay was associated with the expression of locus of enterocyte effacement (LEE)-encoded virulence factors (i.e., intimin and EspD). Indeed, the insertion of a plasmid containing perABC, a transcriptional regulator of virulence factors involved in A/E formation, into aEPEC strains increased and accelerated the formation of A/E lesions. Interestingly, the enhanced expression and translocation of LEE-encoded proteins, such as those expressed in LEE5 (intimin) and LEE4 (EspD), in aEPEC (perABC) was independent of bacterial adhesion. The secretion kinetics of these two proteins representing LEE5 and LEE4 expression correlated with A/E lesion formation. We conclude that the lack of Per in the regulation network of virulence genes is one of the main factors that delay the establishment of A/E lesions induced by aEPEC strains.


Assuntos
Adesinas Bacterianas/metabolismo , Aderência Bacteriana , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Adesinas Bacterianas/genética , Escherichia coli Enteropatogênica/genética , Proteínas de Escherichia coli/genética , Células Hep G2 , Hepatócitos/microbiologia , Humanos , Plasmídeos , Proteínas Repressoras/genética , Fatores de Transcrição/genética
8.
Biomed Res Int ; 2014: 845147, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24883330

RESUMO

The aim of this study was to determine the capacity of biofilm formation of atypical enteropathogenic Escherichia coli (aEPEC) strains on abiotic and biotic surfaces. Ninety-one aEPEC strains, isolated from feces of children with diarrhea, were analyzed by the crystal violet (CV) assay on an abiotic surface after 24 h of incubation. aEPEC strains representing each HEp-2 cell type of adherence were analyzed after 24 h and 6, 12, and 18 days of incubation at 37°C on abiotic and cell surfaces by CFU/cm(2) counting and confocal laser scanning microscopy (CLSM). Biofilm formation on abiotic surfaces occurred in 55 (60.4%) of the aEPEC strains. There was no significant difference in biofilm biomass formation on an abiotic versus prefixed cell surface. The biofilms could be visualized by CLSM at various developmental stages. aEPEC strains are able to form biofilm on an abiotic surface with no association with their adherence pattern on HEp-2 cells with the exception of the strains expressing UND (undetermined adherence). This study revealed the capacity of adhesion and biofilm formation by aEPEC strains on abiotic and biotic surfaces, possibly playing a role in pathogenesis, mainly in cases of persistent diarrhea.


Assuntos
Aderência Bacteriana , Biofilmes/crescimento & desenvolvimento , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Infecções por Escherichia coli/microbiologia , Escherichia coli Enteropatogênica/patogenicidade , Células Epiteliais/citologia , Infecções por Escherichia coli/patologia , Células Hep G2 , Humanos , Receptores Artificiais/química , Propriedades de Superfície
9.
Proc Natl Acad Sci U S A ; 110(7): 2629-34, 2013 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-23359678

RESUMO

Bacterial biofilm formation is a complex developmental process involving cellular differentiation and the formation of intricate 3D structures. Here we demonstrate that exposure to ferric chloride triggers rugose biofilm formation by the uropathogenic Escherichia coli strain UTI89 and by enteric bacteria Citrobacter koseri and Salmonella enterica serovar typhimurium. Two unique and separable cellular populations emerge in iron-triggered, rugose biofilms. Bacteria at the air-biofilm interface express high levels of the biofilm regulator csgD, the cellulose activator adrA, and the curli subunit operon csgBAC. Bacteria in the interior of rugose biofilms express low levels of csgD and undetectable levels of matrix components curli and cellulose. Iron activation of rugose biofilms is linked to oxidative stress. Superoxide generation, either through addition of phenazine methosulfate or by deletion of sodA and sodB, stimulates rugose biofilm formation in the absence of high iron. Additionally, overexpression of Mn-superoxide dismutase, which can mitigate iron-derived reactive oxygen stress, decreases biofilm formation in a WT strain upon iron exposure. Not only does reactive oxygen stress promote rugose biofilm formation, but bacteria in the rugose biofilms display increased resistance to H(2)O(2) toxicity. Altogether, we demonstrate that iron and superoxide stress trigger rugose biofilm formation in UTI89. Rugose biofilm development involves the elaboration of two distinct bacterial populations and increased resistance to oxidative stress.


Assuntos
Biofilmes/crescimento & desenvolvimento , Cloretos/farmacologia , Citrobacter koseri/crescimento & desenvolvimento , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Compostos Férricos/farmacologia , Salmonella typhimurium/crescimento & desenvolvimento , Biofilmes/efeitos dos fármacos , Western Blotting , Citrobacter koseri/efeitos dos fármacos , Escherichia coli Enteropatogênica/efeitos dos fármacos , Proteínas de Escherichia coli/metabolismo , Peróxido de Hidrogênio/metabolismo , Microscopia Confocal , Oxigênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Salmonella typhimurium/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Transativadores/metabolismo , beta-Galactosidase/metabolismo
10.
Curr Biol ; 22(7): 627-31, 2012 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-22386311

RESUMO

The continuous monolayer of intestinal epithelial cells (IECs) lining the gut lumen functions as the site of nutrient absorption and as a physical barrier to prevent the translocation of microbes and associated toxic compounds into the peripheral vasculature. IECs also express host defense proteins such as intestinal alkaline phosphatase (IAP), which detoxify bacterial products and prevent intestinal inflammation. Our laboratory recently showed that IAP is enriched on vesicles that are released from the tips of IEC microvilli and accumulate in the intestinal lumen. Here, we show that these native "lumenal vesicles" (LVs) (1) contain catalytically active IAP that can dephosphorylate lipopolysaccharide (LPS), (2) cluster on the surface of native lumenal bacteria, (3) prevent the adherence of enteropathogenic E. coli (EPEC) to epithelial monolayers, and (4) limit bacterial population growth. We also find that IECs upregulate LV production in response to EPEC and other Gram-negative pathogens. Together, these results suggest that microvillar vesicle shedding represents a novel mechanism for distributing host defense machinery into the intestinal lumen and that microvillus-derived LVs modulate epithelial-microbial interactions.


Assuntos
Fosfatase Alcalina/metabolismo , Vesículas Citoplasmáticas/metabolismo , Escherichia coli Enteropatogênica/imunologia , Células Epiteliais/imunologia , Lipopolissacarídeos/metabolismo , Microvilosidades/metabolismo , Animais , Células CACO-2 , Vesículas Citoplasmáticas/microbiologia , Vesículas Citoplasmáticas/ultraestrutura , Enterócitos/citologia , Enterócitos/metabolismo , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Escherichia coli Enteropatogênica/metabolismo , Humanos , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Microscopia Eletrônica de Transmissão , Microvilosidades/microbiologia , Microvilosidades/ultraestrutura , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo I/metabolismo , Ratos
11.
FEMS Immunol Med Microbiol ; 57(3): 214-28, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19751218

RESUMO

Enteropathogenic E. coli (EPEC) is a common cause of diarrhea in children in developing countries. After adhering to intestinal cells, EPEC secretes effector proteins into host cells, causing cell damage and eventually death. We previously showed that EPEC infection triggers the release of ATP from host cells and that ATP is broken down to ADP, AMP, and adenosine. Adenosine produced from the breakdown of extracellular ATP triggers fluid secretion in intestinal monolayers and may be an important mediator of EPEC-induced diarrhea. Here we examined whether adenosine has any effects on EPEC bacteria. Adenosine stimulated EPEC growth in several types of media in vitro. Adenosine also altered the pattern of EPEC adherence to cultured cells from a localized adherence pattern to a more diffuse pattern. Adenosine changed the expression of virulence factors in EPEC, inhibiting the expression of the bundle-forming pilus (BFP) and enhancing expression of the EPEC secreted proteins (Esps). In vivo, experimental manipulations of adenosine levels had strong effects on the outcome of EPEC infection in rabbit intestinal loops. In addition to its previously reported effects on host tissues, adenosine has strong effects on EPEC bacteria, stimulating EPEC growth, altering its adherence pattern, and changing the expression of several important virulence genes. Adenosine, like noradrenaline, is a small, host-derived molecule that is utilized as a signal by EPEC.


Assuntos
Adenosina/metabolismo , Escherichia coli Enteropatogênica/efeitos dos fármacos , Escherichia coli Enteropatogênica/fisiologia , Retroalimentação Fisiológica , Animais , Aderência Bacteriana/efeitos dos fármacos , Linhagem Celular , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Escherichia coli Enteropatogênica/patogenicidade , Proteínas de Escherichia coli/biossíntese , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Humanos , Íleo/microbiologia , Coelhos , Fatores de Virulência/biossíntese
12.
Cell Microbiol ; 11(3): 521-30, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19134113

RESUMO

In vitro organ culture (IVOC) represents a gold standard model to study enteropathogenic E. coli (EPEC) infection of human intestinal mucosa. However, the optimal examination of the bacterial-host cell interaction requires a directional epithelial exposure, without serosal or cut surface stimulation. A polarized IVOC system (pIVOC) was developed in order to overcome such limitations: apical EPEC infection produced negligible bacterial leakage via biopsy edges, resulted in enhanced colonization compared with standard IVOC, and showed evidence of bacterial detachment, as in natural rabbit EPEC infections. Examination of mucosal innate immune responses in pIVOC showed both interleukin (IL)-8 mRNA and protein levels were significantly increased after apical EPEC infection. Increased IL-8 levels mainly depended on flagellin expression as fliC-negative EPEC did not elicit a significant IL-8 response despite increased mucosal colonization compared with wild-type EPEC. In addition, apical application of purified flagella significantly increased IL-8 protein levels over non-infected controls. Immunofluorescence staining of EPEC-infected small intestinal biopsies revealed apical and basolateral distribution of Toll-like receptor (TLR) 5 on epithelium, suggesting that EPEC can trigger mucosal IL-8 responses by apical flagellin/TLR5 interaction ex vivo and does not require access to the basolateral membrane as postulated in cell culture models.


Assuntos
Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Mucosa Intestinal/microbiologia , Escherichia coli Enteropatogênica/imunologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/imunologia , Flagelina , Deleção de Genes , Perfilação da Expressão Gênica , Humanos , Interleucina-8/biossíntese , Mucosa Intestinal/patologia , Técnicas de Cultura de Órgãos/métodos , Receptor 5 Toll-Like/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA