Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Clin Microbiol Rev ; 27(4): 823-69, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25278576

RESUMO

The pathogenicity and clinical pertinence of diffusely adhering Escherichia coli expressing the Afa/Dr adhesins (Afa/Dr DAEC) in urinary tract infections (UTIs) and pregnancy complications are well established. In contrast, the implication of intestinal Afa/Dr DAEC in diarrhea is still under debate. These strains are age dependently involved in diarrhea in children, are apparently not involved in diarrhea in adults, and can also be asymptomatic intestinal microbiota strains in children and adult. This comprehensive review analyzes the epidemiology and diagnosis and highlights recent progress which has improved the understanding of Afa/Dr DAEC pathogenesis. Here, I summarize the roles of Afa/Dr DAEC virulence factors, including Afa/Dr adhesins, flagella, Sat toxin, and pks island products, in the development of specific mechanisms of pathogenicity. In intestinal epithelial polarized cells, the Afa/Dr adhesins trigger cell membrane receptor clustering and activation of the linked cell signaling pathways, promote structural and functional cell lesions and injuries in intestinal barrier, induce proinflammatory responses, create angiogenesis, instigate epithelial-mesenchymal transition-like events, and lead to pks-dependent DNA damage. UTI-associated Afa/Dr DAEC strains, following adhesin-membrane receptor cell interactions and activation of associated lipid raft-dependent cell signaling pathways, internalize in a microtubule-dependent manner within urinary tract epithelial cells, develop a particular intracellular lifestyle, and trigger a toxin-dependent cell detachment. In response to Afa/Dr DAEC infection, the host epithelial cells generate antibacterial defense responses. Finally, I discuss a hypothetical role of intestinal Afa/Dr DAEC strains that can act as "silent pathogens" with the capacity to emerge as "pathobionts" for the development of inflammatory bowel disease and intestinal carcinogenesis.


Assuntos
Adesinas de Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Escherichia coli/genética , Escherichia coli/patogenicidade , Regulação Bacteriana da Expressão Gênica , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/epidemiologia , Infecções por Escherichia coli/prevenção & controle , Infecções por Escherichia coli/transmissão , Interações Hospedeiro-Patógeno , Humanos , Fatores de Risco , Fatores de Virulência/genética
2.
Clin Microbiol Rev ; 27(2): 167-99, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24696432

RESUMO

A vast and diverse array of microbial species displaying great phylogenic, genomic, and metabolic diversity have colonized the gastrointestinal tract. Resident microbes play a beneficial role by regulating the intestinal immune system, stimulating the maturation of host tissues, and playing a variety of roles in nutrition and in host resistance to gastric and enteric bacterial pathogens. The mechanisms by which the resident microbial species combat gastrointestinal pathogens are complex and include competitive metabolic interactions and the production of antimicrobial molecules. The human intestinal microbiota is a source from which Lactobacillus probiotic strains have often been isolated. Only six probiotic Lactobacillus strains isolated from human intestinal microbiota, i.e., L. rhamnosus GG, L. casei Shirota YIT9029, L. casei DN-114 001, L. johnsonii NCC 533, L. acidophilus LB, and L. reuteri DSM 17938, have been well characterized with regard to their potential antimicrobial effects against the major gastric and enteric bacterial pathogens and rotavirus. In this review, we describe the current knowledge concerning the experimental antibacterial activities, including antibiotic-like and cell-regulating activities, and therapeutic effects demonstrated in well-conducted, placebo-controlled, randomized clinical trials of these probiotic Lactobacillus strains. What is known about the antimicrobial activities supported by the molecules secreted by such probiotic Lactobacillus strains suggests that they constitute a promising new source for the development of innovative anti-infectious agents that act luminally and intracellularly in the gastrointestinal tract.


Assuntos
Antibiose , Terapia Biológica/métodos , Gastroenteropatias/terapia , Trato Gastrointestinal/microbiologia , Lactobacillus/fisiologia , Probióticos , Humanos , Lactobacillus/crescimento & desenvolvimento , Ensaios Clínicos Controlados Aleatórios como Assunto
3.
Microbiology (Reading) ; 159(Pt 9): 1956-1971, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23873784

RESUMO

We conducted experiments in order to examine whether the probiotic Lactobacillus casei strain Shirota YIT9029 (LcS) in vitro and in vivo antagonism of Helicobacter pylori and Salmonella, involves inhibition of the swimming motility of these pathogens. We report the irreversible inhibition of the swimming motility of H. pylori strain 1101 and reversible inhibition of Salmonella enterica serovar Typhimurium (S. Typhimurium) strain SL1344 by compound(s) secreted by LcS. In H. pylori 1101, irreversible inhibition results in the helical cells being progressively replaced by cells with 'c'-shaped and coccoid morphologies, accompanied by a loss of FlaA and FlaB flagellin expression. In S. Typhimurium SL1344, transient inhibition develops after membrane depolarization and without modification of expression of FliC flagellin. The inhibitory activity of strain LcS against both S. Typhimurium and H. pylori swimming motilities is linked with a small sized, heat-sensitive, and partially trypsin-sensitive, secreted compound(s), and needed the cooperation of the secreted membrane permeabilizing lactic acid metabolite. The inhibition of S. Typhimurium SL1344 swimming motility leads to delayed cell entry into human enterocyte-like Caco-2/TC7 cells and a strong decrease of cell entry into human mucus-secreting HT29-MTX cells.


Assuntos
Antibiose , Fatores Biológicos/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/fisiologia , Lacticaseibacillus casei/fisiologia , Infecções por Salmonella/microbiologia , Salmonella typhimurium/fisiologia , Proteínas de Bactérias/metabolismo , Fatores Biológicos/farmacologia , Linhagem Celular , Meios de Cultura/química , Meios de Cultura/metabolismo , Flagelina/metabolismo , Helicobacter pylori/efeitos dos fármacos , Humanos , Lacticaseibacillus casei/química , Salmonella typhimurium/efeitos dos fármacos
4.
Infect Immun ; 80(5): 1891-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22371374

RESUMO

We recently documented the neutrophil response to enterovirulent diffusely adherent Escherichia coli expressing Afa/Dr fimbriae (Afa/Dr DAEC), using the human myeloid cell line PLB-985 differentiated into fully mature neutrophils. Upon activation, particularly during infections, neutrophils release neutrophil extracellular traps (NETs), composed of a nuclear DNA backbone associated with antimicrobial peptides, histones, and proteases, which entrap and kill pathogens. Here, using fluorescence microscopy and field emission scanning electron microscopy, we observed NET production by PLB-985 cells infected with the Afa/Dr wild-type (WT) E. coli strain C1845. We found that these NETs were able to capture, immobilize, and kill WT C1845 bacteria. We also developed a coculture model of human enterocyte-like Caco-2/TC7 cells and PLB-985 cells previously treated with WT C1845 and found, for the first time, that the F-actin cytoskeleton of enterocyte-like cells is damaged in the presence of bacterium-induced NETs and that this deleterious effect is prevented by inhibition of protease release. These findings provide new insights into the neutrophil response to bacterial infection via the production of bactericidal NETs and suggest that NETs may damage the intestinal epithelium, particularly in situations such as inflammatory bowel diseases.


Assuntos
Adesinas de Escherichia coli/metabolismo , Enterócitos/citologia , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Neutrófilos/fisiologia , alfa-Defensinas/metabolismo , Adesinas de Escherichia coli/genética , Linhagem Celular , Técnicas de Cocultura , Escherichia coli/ultraestrutura , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Histonas/metabolismo , Humanos , Peptídeo Hidrolases/metabolismo
5.
Cell Microbiol ; 13(7): 992-1013, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21501364

RESUMO

The secreted autotransporter toxin, Sat, which belongs to the subfamily of serine protease autotransporters of Enterobacteriaceae, acts as a virulence factor in extraintestinal and intestinal pathogenic strains of Escherichia coli. We observed that HeLa cells exposed to the cell-free culture supernatant of recombinant strain AAEC185p(Sat-IH11128) producing the Sat toxin (CFCS(Sat) ), displayed dramatic disorganization of the F-actin cytoskeleton before loosening cell-to-cell junctions and detachment. Examination of the effect of Sat on GFP-microtubule-associated protein light chain 3 (LC3) HeLa cells revealed that CFCS(Sat) -induced autophagy follows CFCS(Sat) -induced F-actin cytoskeleton rearrangement. The induced autophagy shows an acceleration of the autophagy flux soon after Sat treatment, followed later by a blockade of the flux leading to the accumulation of large GFP-LC3-positive vacuoles in the cell cytoplasm. CFCS(Sat) did not induce cell detachment in autophagy-deficient mouse embryonic fibroblasts in contrast with wild-type mouse embryonic fibroblasts. The CFCS(Sat) -induced large GFP-LC3 dots do not display the characteristics of autophagolysosomes including expression of cathepsin D and Lamp-1 and 2 proteins, and Lysotracker Red- and DQ-BSA-positive labelling. We provide evidences that CFCS(Sat) -induced autophagy is not a cell response intended to get rid of the intracellular toxin. By a pharmacological blockers approach, we found that the blockade of Erk1/2 and p38 MAPKs, but not JNK, inhibited the CFCS(Sat) -induced autophagy and cell detachment whereas phosphatidylinositol-3 kinase blockers inhibiting canonical autophagy were inactive. When attached CFCS(Sat) -treated cells start to detach they showed caspase-independent cell death and rearrangements of the focal adhesion-associated vinculin and paxillin. Collectively, our results support that Sat triggers autophagy in epithelial cells that relies on its cell-detachment effect.


Assuntos
Autofagia , Adesão Celular , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Proteínas de Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Fatores de Virulência/metabolismo , Animais , Toxinas Bacterianas , Citoesqueleto/metabolismo , Fibroblastos/microbiologia , Células HeLa , Humanos , Camundongos , Transdução de Sinais
6.
Cell Microbiol ; 13(5): 764-85, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21352462

RESUMO

CEACAM1 expressed by granulocytes and epithelial cells is recognized as a membrane-associated receptor by some Gram-negative pathogens. Here we report a previously unsuspected role of human CEACAM1-4L (hCEACAM1-4L) in polarized epithelial cells. We find that in contrast with non-transfected cells, Madin Darby Canine Kidney strain II (MDCK) engineered for the apical expression of the long cytoplasmic chain protein hCEACAM1-4L showed a serum-independent increase in the phosphorylation of the extracellular signal-regulated kinase 1/2 (Erk1/2) and p38 mitogen-activated protein kinases (MAPKs) after treatment with lipopolysaccharide (LPS) of wild-type, diffusely adhering Afa/Dr Escherichia coli (Afa/Dr DAEC) strain IH11128. Aggregates of FITC-LPS bind the apical domain of MDCK-hCEACAM1-4L cells colocalizing with the apically expressed hCEACAM1-4L protein and do not bind MDCK-pCEP cells, and surface plasmon resonance analysis shows that LPS binds to the extracellular domain of the CEACAM1-4L protein. We showed that cell polarization and lipid rafts positively control the LPS-IH11128-induced phosphorylation of Erk1/2 in MDCK-hCEACAM1-4L cells. Structure-function analysis using mutated hCEACAM1-4L protein shows that the cytoplasmic domain of the protein is needed for LPS-induced MAPK signalling, and that phosphorylation of Tyr-residues is not increased in association with MAPK signalling. The hCEACAM1-4L-dependent Erk1/2 phosphorylation develops in the presence of lipid A and does not develop in the presence of penta-acylated LPS. Finally, small interfering RNA (siRNA) silencing of canine TLR4 abolishes the hCEACAM1-4L-dependent, LPS-induced phosphorylation of Erk1/2. Collectively, our results support the notion that the apically expressed, full-length hCEACAM1-4L protein functions as a novel LPS-conveying molecule at the mucosal surface of polarized epithelial cells for subsequent MD-2/TLR4 receptor-dependent MAPK Erk1/2 and p38 signalling.


Assuntos
Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Rim/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Antígenos CD/genética , Moléculas de Adesão Celular/genética , Linhagem Celular , Polaridade Celular , Cães , Escherichia coli/química , Engenharia Genética , Humanos , Lipídeo A , Lipopolissacarídeos/imunologia , Microdomínios da Membrana/metabolismo , Mucosa/metabolismo , Mucosa/fisiologia , Fosforilação , Isoformas de Proteínas/genética , Interferência de RNA , RNA Interferente Pequeno , Ressonância de Plasmônio de Superfície , Receptor 4 Toll-Like/genética
7.
Antimicrob Agents Chemother ; 55(10): 4810-20, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21825295

RESUMO

We report that both culture and the cell-free culture supernatant (CFCS) of Lactobacillus acidophilus strain LB (Lactéol Boucard) have the ability (i) to delay the appearance of Salmonella enterica serovar Typhimurium strain SL1344-induced mobilization of F-actin and, subsequently, (ii) to retard cell entry by S. Typhimurium SL1344. Time-lapse imaging and Western immunoblotting showed that S. Typhimurium SL1344 swimming motility, as represented by cell tracks of various types, was rapidly but temporarily blocked without affecting the expression of FliC flagellar propeller protein. We show that the product(s) secreted by L. acidophilus LB that supports the inhibitory activity is heat stable and of low molecular weight. The product(s) caused rapid depolarization of the S. Typhimurium SL1344 cytoplasmic membrane without affecting bacterial viability. We identified inhibition of swimming motility as a newly discovered mechanism by which the secreted product(s) of L. acidophilus strain LB retards the internalization of the diarrhea-associated pathogen S. enterica serovar Typhimurium within cultured human enterocyte-like cells.


Assuntos
Enterócitos/microbiologia , Flagelos/fisiologia , Lactobacillus acidophilus/fisiologia , Probióticos , Salmonella typhimurium/fisiologia , Actinas/antagonistas & inibidores , Actinas/metabolismo , Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Células CACO-2 , Membrana Celular/fisiologia , Células Cultivadas , Humanos , Viabilidade Microbiana , Movimento , Salmonella typhimurium/patogenicidade , Imagem com Lapso de Tempo
8.
Infect Immun ; 78(7): 2974-83, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20404079

RESUMO

The enterovirulent Escherichia coli strains potentially involved in inflammatory bowel diseases include diffusely adherent strains expressing Afa/Dr fimbriae (Afa/Dr DAEC). We have previously observed type 1 pilus-mediated interleukin-8 (IL-8) hyperproduction in infected neutrophils. As pathogen induction of host cell death programs and clearance of apoptotic infected cells are crucial for innate immune system homeostasis and host integrity, we examined modulation of neutrophil cell death by Afa/Dr DAEC. Using the human PLB-985 cell line differentiated into fully mature neutrophils, we found that the wild-type enterovirulent E. coli strain C1845 and the recombinant strain DH5alpha/pF1845 (expressing the fimbrial adhesin F1845) similarly induced time-dependent phosphatidylserine (PS) externalization, suggesting a major specific role of this virulence factor. Using small interfering RNA (siRNA) decay-accelerating factor (DAF)-transfected PLB-985 cells, we then showed that this PS externalization was triggered in part by glycosylphosphatidylinositol (GPI)-anchored DAF receptor engagement (leading to tyrosine kinase and protein kinase C activation) and that it required cytoskeleton and lipid raft architectural integrity. PS externalization under these conditions was not dependent on caspases, mitochondria, lysosomes, or reactive oxygen or nitrogen species. F1845-mediated PS externalization was sufficient to enable macrophage engulfment of infected differentiated PLB-985 cells. These findings provide new insights into the neutrophil response to Afa/Dr DAEC infection and highlight a new role for F1845 fimbriae. Interestingly, although apoptosis pathways were not engaged, C1845-infected PLB-985 cells displayed enhanced removal by macrophages, a process that may participate in the resolution of Afa/Dr DAEC infection and related inflammation.


Assuntos
Escherichia coli Enteropatogênica/fisiologia , Proteínas de Escherichia coli/fisiologia , Fímbrias Bacterianas/fisiologia , Fatores de Transcrição/fisiologia , Adesinas de Escherichia coli/fisiologia , Apoptose/fisiologia , Aderência Bacteriana/fisiologia , Western Blotting , Linhagem Celular Tumoral , Infecções por Escherichia coli/microbiologia , Proteínas de Fímbrias/fisiologia , Granulócitos/microbiologia , Granulócitos/fisiologia , Humanos , Lisossomos/microbiologia , Lisossomos/fisiologia , Macrófagos/microbiologia , Macrófagos/fisiologia , Neutrófilos/microbiologia , Neutrófilos/fisiologia , Fagocitose/fisiologia
9.
Infect Immun ; 78(3): 927-38, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20065027

RESUMO

In rabbit ligated ileal loops, two atypical enteropathogenic Escherichia coli (aEPEC) strains, 3991-1 and 0421-1, intimately associated with the cell membrane, forming the characteristic EPEC attachment and effacement lesion of the brush border, induced a mucous hypersecretion, whereas typical EPEC (tEPEC) strain E2348/69 did not. Using cultured human mucin-secreting intestinal HT29-MTX cells, we demonstrate that apically aEPEC infection is followed by increased production of secreted MUC2 and MUC5AC mucins and membrane-bound MUC3 and MUC4 mucins. The transcription of the MUC5AC and MUC4 genes was transiently upregulated after aEPEC infection. We provide evidence that the apically adhering aEPEC cells exploit the mucins' increased production since they grew in the presence of membrane-bound mucins, whereas tEPEC did not. The data described herein report a putative new virulence phenomenon in aEPEC.


Assuntos
Aderência Bacteriana , Enterócitos/metabolismo , Enterócitos/microbiologia , Escherichia coli Enteropatogênica/patogenicidade , Mucinas/biossíntese , Animais , Linhagem Celular , Modelos Animais de Doenças , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Infecções por Escherichia coli/microbiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Íleo/microbiologia , Íleo/patologia , Coelhos , Regulação para Cima , Virulência
10.
J Leukoc Biol ; 85(2): 310-21, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19015376

RESUMO

The innate immune response to enteropathogenic bacteria includes chemokine-induced polymorphonuclear neutrophil (PMN) migration across mucosal epithelia leading to bacterial clearance and resolution of infection. Among these bacteria, diffusely adherent Escherichia coli expressing Afa/Dr fimbriae (Afa/Dr DAEC), causing childhood diarrhea, can promote IL-8-dependent PMN transmigration across cultured intestinal epithelial cell monolayers via MAPK pathway activation. However, interactions between PMN and Afa/Dr DAEC are poorly documented and constitute the aim of the present study. Using the human PLB-985 cell line differentiated into fully mature PMN, we described the coordinated response to various E. coli. The rapid and strong release of reactive oxygen species and preformed intragranular mediators (myeloperoxidase and IL-8) is followed by a later TNF-alpha, IL-1beta, and IL-8 synthesis. The use of wild-type (IH11128, C1845, LF82), control (AAEC185), and recombinant (AAEC185 bearing Dr or F1845 fimbriae, AdLF82, or type 1 pili) bacterial strains allowed us to demonstrate that late IL-8 hyperproduction is triggered by type 1 pili but not by Dr or F1845 fimbriae; MAPKs (p38, ERK, Src) and NF-kappaB activations are implicated in this response. Thus, in the course of Afa/Dr DAEC intestinal infection, epithelium- and neutrophil-derived IL-8 could, at least in part, control the flow of neutrophils through the lamina propria. Afa/Dr DAEC-induced IL-8 hyperproduction by PMN might thus be important for inducing and perpetuating local inflammation, and this self-amplifying loop might play a role in the pathogenesis of inflammatory bowel diseases such as Crohn's disease.


Assuntos
Diferenciação Celular , Escherichia coli/imunologia , Fímbrias Bacterianas/imunologia , Interleucina-8/biossíntese , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neutrófilos/enzimologia , Quinases da Família src/metabolismo , Antígenos CD/imunologia , Aderência Bacteriana , Antígeno CD11b/imunologia , Antígenos CD18/imunologia , Moléculas de Adesão Celular/imunologia , Escherichia coli/citologia , Proteínas Ligadas por GPI , Humanos , Lipopolissacarídeos/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Neutrófilos/citologia , Neutrófilos/microbiologia , Peroxidase/metabolismo , Explosão Respiratória/imunologia , Quinases da Família src/antagonistas & inibidores
11.
Infect Immun ; 77(1): 517-31, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19015254

RESUMO

We used transfected epithelial CHO-B2 cells as a model to identify the mechanism mediating internalization of Afa/Dr diffusely adhering Escherichia coli. We provide evidence that neither the alpha5 or beta1 integrin subunits nor alpha5beta1 integrin functioned as a receptor mediating the adhesion and/or internalization of Dr or Afa-III fimbria-positive bacteria. We also demonstrated that (i) whether or not the AfaD or DraD invasin subunits were present, there was no difference in the cell association and entry of bacteria and that (ii) DraE or AfaE-III adhesin subunits are necessary and sufficient to promote the receptor-mediated bacterial internalization into epithelial cells expressing human decay-accelerating factor (DAF), CEACAM1, CEA, or CEACAM6. Internalization of Dr fimbria-positive E. coli within CHO-DAF, CHO-CEACAM1, CHO-CEA, or CHO-CEACAM6 cells occurs through a microfilament-independent, microtubule-dependent, and lipid raft-dependent mechanism. Wild-type Dr fimbria-positive bacteria survived better within cells expressing DAF than bacteria internalized within CHO-CEACAM1, CHO-CEA, or CHO-CEACAM6 cells. In DAF-positive cells, internalized Dr fimbria-positive bacteria were located in vacuoles that contained more than one bacterium, displaying some of the features of late endosomes, including the presence of Lamp-1 and Lamp-2, and some of the features of CD63 proteins, but not of cathepsin D, and were acidic. No interaction between Dr fimbria-positive-bacterium-containing vacuoles and the autophagic pathway was observed.


Assuntos
Aderência Bacteriana , Citoplasma/microbiologia , Escherichia coli/fisiologia , Receptores de Superfície Celular/fisiologia , Adesinas de Escherichia coli/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos CD55/metabolismo , Células CHO , Moléculas de Adesão Celular/metabolismo , Cricetinae , Cricetulus , Células Epiteliais/microbiologia , Proteínas Ligadas por GPI , Células HeLa , Humanos , Viabilidade Microbiana , Receptores de Superfície Celular/metabolismo , Vacúolos/microbiologia
12.
Cell Microbiol ; 10(3): 632-54, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17979980

RESUMO

Human decay accelerating factor (hDAF, CD55) and members of the carcinoembryonic-antigen-related cell-adhesion molecules (hCEACAMs) family are recognized as receptors by Gram-negative, diffusely adhering Escherichia coli (DAEC) strains expressing Afa/Dr adhesins. We report here that hCEACAM1-4L has a key function in downregulating the protein tyrosine Src kinase associated with hDAF signalling. After infecting HeLa epithelial cells stably transfected with hCEACAM1-4L cDNA with Dr adhesin-positive E. coli, the amount of the pTyr(416)-active form of the Src protein decreased, whereas that of the pTyr(527)-inactive form of Src protein did not increase. This downregulation of the Src protein implies that part of the hCEACAM1-4L protein had been translocated into lipid rafts, the protein was phosphorylated at Tyr residues in the cytoplasmic domain, and it was physically associated with the protein tyrosine phosphatase, SHP-2. Finally, we found that the hCEACAM1-4L-associated SHP-2 was not phosphorylated and lacked phosphatase activity, suggesting that the downregulation of Src protein associated with hDAF signalling results from the absence of dephosphorylation of the pTyr(527)-inactive form necessary for Src kinase activation.


Assuntos
Antígenos CD/metabolismo , Antígenos CD55/metabolismo , Moléculas de Adesão Celular/metabolismo , Escherichia coli/fisiologia , Quinases da Família src/metabolismo , Adesinas de Escherichia coli/metabolismo , Regulação para Baixo , Células HeLa , Humanos , Microdomínios da Membrana/química , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo
13.
Microbes Infect ; 8(7): 1851-8, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16815721

RESUMO

Afa/Dr diffusely adhering Escherichia coli have been shown to cause urinary tract infections and enteric infections. Virulence of Dr-positive IH11128 bacteria is associated with the presence of Dr fimbriae. In this report, we show for the first time that the Dr fimbriae are released in the extracellular medium in response to multiple environmental signals. Production and secretion of Dr fimbriae are clearly thermoregulated. A comparison of the amounts of secreted fimbriae showed that the secretion is drastically increased during anaerobic growth in minimal medium. The effect of anaerobiosis on secretion seemed to depend on both the growth phase and the culture medium. The secretion was maximal during the logarithmic-phase growth and corresponded to 27 and 57% of total Dr fimbriae produced by bacteria grown in mineral medium+glucose and LB broth, respectively. Thus, the anaerobic environment of the colon would favour the secretion of Dr fimbriae during bacterial multiplication. The controlled release of the Dr fimbriae, which is carried out in the absence of cellular lysis, appears independent of the action of proteases or a process of maturation. The mechanism employed in the liberation of Dr fimbriae thus seems different from that described for the adhesins FHA and Hap of Bordetella pertussis and Haemophilus influenzae.


Assuntos
Escherichia coli/metabolismo , Fímbrias Bacterianas/metabolismo , Fatores de Virulência/metabolismo , Adesinas de Escherichia coli/metabolismo , Anaerobiose , Aderência Bacteriana , Células CACO-2 , Meios de Cultura/química , Humanos , Transporte Proteico , Temperatura
14.
Res Microbiol ; 157(3): 241-7, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16266797

RESUMO

Six Lactobacillus strains including commercial probiotic ones (L. acidophilus IBB 801, L. amylovorus DCE 471, L. casei Shirota, L. johnsonii La1, L. plantarum ACA-DC 287 and L. rhamnosus GG) were investigated, through batch fermentations under controlled conditions, for their capacity to inhibit Salmonella enterica serovar Typhimurium SL1344. All lactobacilli displayed strong antibacterial activity toward this Gram-negative pathogen and significantly inhibited invasion of the pathogen into cultured human enterocyte-like Caco-2/TC7 cells. By studying the production kinetics of antibacterial activity and applying the appropriate acid and pH control samples during a killing assay, we were able to distinguish between the effect of lactic acid and other inhibitory compounds produced. The antibacterial activity of L. acidophilus IBB 801, L. amylovorus DCE 471, L. casei Shirota and L. rhamnosus GG was solely due to the production of lactic acid. The antibacterial activity of L. johnsonii La1 and L. plantarum ACA-DC 287 was due to the production of lactic acid and (an) unknown inhibitory substance(s). The latter was (were) only active in the presence of lactic acid. In addition, the lactic acid produced was responsible for significant inhibitory activity upon invasion of Salmonella into Caco-2/TC7 cells.


Assuntos
Ácido Láctico/metabolismo , Lactobacillus/fisiologia , Probióticos/metabolismo , Salmonella typhimurium/fisiologia , Células CACO-2 , Fermentação , Humanos , Concentração de Íons de Hidrogênio , Lactobacillus/crescimento & desenvolvimento , Lactobacillus/metabolismo , Viabilidade Microbiana , Salmonella typhimurium/crescimento & desenvolvimento
15.
FEMS Microbiol Lett ; 256(2): 185-94, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16499605

RESUMO

Diffusely adherent Escherichia coli (DAEC) strains are currently considered to constitute a putative sixth group of diarrheagenic E. coli. However, on the basis of their diffuse adherence to HEp-2 and HeLa cells, the detection of afa/dra/daa-related operons encoding this adherence phenotype, and the mobilization of decay-accelerating factor, both commensal and pathogenic strains can be classified as Afa/Dr DAEC isolates. Furthermore, strains associated with diarrheal diseases and strains causing extra-intestinal infections can also be identified as Afa/Dr DAEC strains. Although several cell signaling events that occur after epithelial cells have been infected by Afa/Dr DAEC have been reported, the pathophysiological processes that allow intestinal and extra-intestinal infections to develop are not fully understood. This review focuses on the genetic organization of the afa/dra/daa-related operons and on the virulence factors that trigger cellular responses, some of which are deleterious for the host cells. Finally, this review suggests future lines of research that could help to elucidate these questions.


Assuntos
Adesinas de Escherichia coli/fisiologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/fisiologia , Escherichia coli/patogenicidade , Fatores de Transcrição/fisiologia , Adesinas de Escherichia coli/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Humanos , Óperon , Fatores de Transcrição/genética , Fatores de Virulência/genética , Fatores de Virulência/fisiologia
16.
FEMS Microbiol Lett ; 257(1): 132-8, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16553843

RESUMO

The purpose of this study was to investigate the antibacterial activities of Lactobacillus jensenii KS119.1 and KS121.1, and Lactobacillus gasserii KS120.1 and KS124.3 strains isolated from the vaginal microflora of healthy women, against uropathogenic, diffusely adhering Afa/Dr Escherichia coli (Afa/Dr DAEC) strains IH11128 and 7372 involved in recurrent cystitis. We observed that some of the Lactobacillus isolates inhibited the growth and decreased the viability of E. coli IH11128 and 7372. In addition, we observed that adhering Lactobacillus strains inhibited adhesion of E. coli IH11128 onto HeLa cells, and inhibited internalization of E. coli IH11128 within HeLa cells.


Assuntos
Antibiose , Infecções por Escherichia coli/microbiologia , Escherichia coli/crescimento & desenvolvimento , Lactobacillus/crescimento & desenvolvimento , Infecções Urinárias/microbiologia , Vagina/microbiologia , Adesinas de Escherichia coli/metabolismo , Aderência Bacteriana , Cistite/microbiologia , Escherichia coli/patogenicidade , Feminino , Células HeLa/microbiologia , Humanos , Lactobacillus/isolamento & purificação
17.
FEMS Immunol Med Microbiol ; 48(3): 424-32, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17059467

RESUMO

The purpose of this study was to investigate how human vaginal isolates of Lactobacillus acidophilus, Lactobacillus jensenii, Lactobacillus gasseri and Lactobacillus crispatus inhibit the vaginosis-associated pathogens Gardnerella vaginalis and Prevotella bivia. Results show that all the strains in coculture condition reduced the viability of G. vaginalis and P. bivia, but with differing degrees of efficacy. The treatment of G. vaginalis- and P. bivia-infected cultured human cervix epithelial HeLa cells with L. gasseri strain KS120.1 culture or cell-free culture supernatant (CFCS) results in the killing of the pathogens that are adhering to the cells. The mechanism of the killing activity is not attributable to low pH and the presence of lactic acid alone, but rather to the presence of hydrogen peroxide and proteolytic enzyme-resistant compound(s) present in the CFCSs. In addition, coculture of G. vaginalis or P. bivia with L. gasseri KS120.1 culture or KS120.1 bacteria results in inhibition of the adhesion of the pathogens onto HeLa cells.


Assuntos
Antibacterianos/metabolismo , Gardnerella vaginalis/crescimento & desenvolvimento , Lactobacillus/metabolismo , Prevotella/crescimento & desenvolvimento , Vagina/microbiologia , Vaginose Bacteriana/microbiologia , Aderência Bacteriana , Técnicas de Cocultura , Feminino , Células HeLa , Humanos , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Ácido Láctico/metabolismo , Ácido Láctico/farmacologia , Vaginose Bacteriana/prevenção & controle
18.
FEMS Microbiol Rev ; 28(4): 405-40, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15374659

RESUMO

The gastrointestinal tract is a complex ecosystem that associates a resident microbiota and cells of various phenotypes lining the epithelial wall expressing complex metabolic activities. The resident microbiota in the digestive tract is a heterogeneous microbial ecosystem containing up to 1 x 10(14) colony-forming units (CFUs) of bacteria. The intestinal microbiota plays an important role in normal gut function and maintaining host health. The host is protected from attack by potentially harmful microbial microorganisms by the physical and chemical barriers created by the gastrointestinal epithelium. The cells lining the gastrointestinal epithelium and the resident microbiota are two partners that properly and/or synergistically function to promote an efficient host system of defence. The gastrointestinal cells that make up the epithelium, provide a physical barrier that protects the host against the unwanted intrusion of microorganisms into the gastrointestinal microbiota, and against the penetration of harmful microorganisms which usurp the cellular molecules and signalling pathways of the host to become pathogenic. One of the basic physiological functions of the resident microbiota is that it functions as a microbial barrier against microbial pathogens. The mechanisms by which the species of the microbiota exert this barrier effect remain largely to be determined. There is increasing evidence that lactobacilli and bifidobacteria, which inhabit the gastrointestinal microbiota, develop antimicrobial activities that participate in the host's gastrointestinal system of defence. The objective of this review is to analyze the in vitro and in vivo experimental and clinical studies in which the antimicrobial activities of selected lactobacilli and bifidobacteria strains have been documented.


Assuntos
Antibiose , Bactérias/crescimento & desenvolvimento , Infecções Bacterianas/prevenção & controle , Bifidobacterium/fisiologia , Gastroenteropatias/prevenção & controle , Trato Gastrointestinal/microbiologia , Lactobacillus/fisiologia , Rotavirus/crescimento & desenvolvimento , Infecções Urinárias/prevenção & controle , Animais , Antibacterianos/biossíntese , Bactérias/patogenicidade , Aderência Bacteriana , Infecções Bacterianas/microbiologia , Gastroenteropatias/microbiologia , Humanos , Peróxido de Hidrogênio/metabolismo , Rotavirus/patogenicidade , Infecções por Rotavirus/prevenção & controle , Infecções por Rotavirus/virologia , Tensoativos/metabolismo , Infecções Urinárias/microbiologia
19.
FEBS J ; 272(17): 4385-97, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16128808

RESUMO

We have defined the in vivo and in vitro metabolic fate of internalized cholera toxin (CT) in the endosomal apparatus of rat liver. In vivo, CT was internalized and accumulated in endosomes where it underwent degradation in a pH-dependent manner. In vitro proteolysis of CT using an endosomal lysate required an acidic pH and was sensitive to pepstatin A, an inhibitor of aspartic acid proteases. By nondenaturating immunoprecipitation, the acidic CT-degrading activity was attributed to the luminal form of endosomal cathepsin D. The rate of toxin hydrolysis using an endosomal lysate or pure cathepsin D was found to be high for native CT and free CT-B subunit, and low for free CT-A subunit. On the basis of IC(50) values, competition studies revealed that CT-A and CT-B subunits share a common binding site on the cathepsin D enzyme, with native CT and free CT-B subunit displaying the highest affinity for the protease. By immunofluorescence, partial colocalization of internalized CT with cathepsin D was confirmed at early times of endocytosis in both hepatoma HepG2 and intestinal Caco-2 cells. Hydrolysates of CT generated at low pH by bovine cathepsin D displayed ADP-ribosyltransferase activity towards exogenous Gsalpha protein suggesting that CT cytotoxicity, at least in part, may be related to proteolytic events within endocytic vesicles. Together, these data identify the endocytic apparatus as a critical subcellular site for the accumulation and proteolytic degradation of endocytosed CT, and define endosomal cathepsin D an enzyme potentially responsible for CT cytotoxic activation.


Assuntos
Catepsina D/metabolismo , Toxina da Cólera/metabolismo , Endossomos/metabolismo , Fígado/metabolismo , Animais , Biodegradação Ambiental , Biotransformação , Células CACO-2 , Bovinos , Linhagem Celular , Toxina da Cólera/química , Endocitose , Humanos , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Cinética , Masculino , Subunidades Proteicas , Ratos , Ratos Sprague-Dawley
20.
Best Pract Res Clin Gastroenterol ; 17(5): 741-54, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14507585

RESUMO

Probiotic lactic acid strains are live micro-organisms that, when consumed in adequate amounts as part of food, confer a health benefit on the host. The scientific basis for the use of selected probiotic strains has only recently been firmly established, and appropriate and well-conducted experimental in vitro and in vivo studies, as well as clinical studies, are now beginning to be published, especially with regard to the effectiveness of probiotic strains in antagonizing pathogens. In particular, experimental data have allowed new insights into selected probiotic strains that express strain-specific probiotic properties and into the mechanism of action of these strains. The objective of this review is to analyse the in vitro or in vivo experimental studies in which the antimicrobial activity of selected Lactobacillus and Bifidobacterium strains has been documented.


Assuntos
Antibiose , Aderência Bacteriana , Bifidobacterium/fisiologia , Mucosa Intestinal/microbiologia , Lactobacillus/fisiologia , Probióticos , Animais , Antibiose/fisiologia , Infecções Bacterianas/microbiologia , Infecções Bacterianas/fisiopatologia , Gastroenteropatias/microbiologia , Gastroenteropatias/fisiopatologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA