Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-31681620

RESUMO

Pet and EspC are toxins secreted by enteroaggregative (EAEC) and enteropathogenic (EPEC) diarrheagenic Escherichia coli pathotypes, respectively. Both toxins are members of the Serine Protease Autotransporters of Enterobacteriaceae (SPATEs) family. Pet and EspC are important virulence factors that produce cytotoxic and enterotoxic effects on enterocytes. Here, we evaluated the effect of curcumin, a polyphenolic compound obtained from the rhizomes of Curcuma longa L. (Zingiberaceae) on the secretion and cytotoxic effects of Pet and EspC proteins. We found that curcumin prevents Pet and EspC secretion without affecting bacterial growth or the expression of pet and espC. Our results show that curcumin affects the release of these SPATEs from the translocation domain, thereby affecting the pathogenesis of EAEC and EPEC. Curcumin-treated EAEC and EPEC did not induce significant cell damage like the ability to disrupt the actin cytoskeleton, without affecting their characteristic adherence patterns on epithelial cells. A molecular model of docking predicted that curcumin interacts with the determinant residues Asp1018-Asp1019 and Asp1029-Asp1030 of the translocation domain required for the release of Pet and EspC, respectively. Consequently, curcumin blocks Pet and EspC cytotoxicity on epithelial cells by preventing their release from the outer membrane.


Assuntos
Membrana Externa Bacteriana/metabolismo , Toxinas Bacterianas/metabolismo , Curcumina/farmacologia , Escherichia coli Enteropatogênica/efeitos dos fármacos , Escherichia coli Enteropatogênica/fisiologia , Enterotoxinas/metabolismo , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/metabolismo , Serina Endopeptidases/metabolismo , Toxinas Bacterianas/química , Sítios de Ligação , Curcumina/química , Citoesqueleto/metabolismo , Enterotoxinas/química , Proteínas de Escherichia coli/química , Interações Hospedeiro-Patógeno , Humanos , Modelos Moleculares , Conformação Molecular , Ligação Proteica , Proteólise , Serina Endopeptidases/química , Relação Estrutura-Atividade
2.
Microbiologyopen ; 8(12): e931, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31568664

RESUMO

Enteropathogenic Escherichia coli (EPEC) infection causes a histopathological lesion including recruitment of F-actin beneath the attached bacteria and formation of actin-rich pedestal-like structures. Another important target of EPEC is the tight junction (TJ), and EspF induces displacement of TJ proteins and increased intestinal permeability. Previously, we determined that an EPEC strain lacking EspF did not cause TJ disruption; meanwhile, pedestals were located on the TJ and smaller than those induced by the wild-type strain. Therefore, EspF could be playing an important role in both phenotypes. Here, using different cell models, we found that EspF was essential for pedestal maturation through ZO-1 disassembly from TJ, leading to (a) ZO-1 recruitment to the pedestal structure; no other main TJ proteins were required. Recruited ZO-1 allowed the afadin recruitment. (b) Afadin recruitment caused an afadin-ZO-1 transient interaction, like during TJ formation. (c) Afadin and ZO-1 were segregated to the tip and the stem of pedestal, respectively, causing pedestal maturation. Initiation of these three discrete phases for pedestal maturation functionally and physically required EspF expression. Pedestal maturation process could help coordinate the epithelial actomyosin function by maintaining the actin-rich column composing the pedestal structure and could be important in the dynamics of the pedestal movement on epithelial cells.


Assuntos
Escherichia coli Enteropatogênica/fisiologia , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas/genética , Junções Íntimas/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo , Actinas/metabolismo , Células Epiteliais/metabolismo , Infecções por Escherichia coli/patologia , Proteínas de Escherichia coli/metabolismo , Imunofluorescência , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Permeabilidade , Fosfoproteínas/metabolismo , Ligação Proteica
3.
Artigo em Inglês | MEDLINE | ID: mdl-27774437

RESUMO

Inflammatory response is key for the host defense against diarrheagenic Escherichia coli and contributes to the pathogenesis of the disease but there is not a comparative study among different diarrheagenic pathotypes. We analyzed the inflammatory response induced by five diarrheagenic pathotypes in a HT-29 cell infection model. The model was unified to reproduce the pathogenesis of each pathotype. To compare the inflammatory responses we evaluated: (i) nuclear NF-κB and ERK1/2 translocation by confocal microscopy; (ii) kinetics of activation by each pathway detecting p65 and ERK1/2 phosphorylation by Western blotting; (iii) pathways modulation through bacterial infections with or without co-stimulation with TNF-α or EGF; (iv) cytokine profile induced by each pathotype with and without inhibitors of each pathway. EHEC but mainly EPEC inhibited translocation and activation of p65 and ERK1/2 pathways, as well as cytokines secretion; inhibition of p65 and ERK1/2 phosphorylation prevailed in the presence of TNF-α and EGF, respectively. Intracellular strains, EIEC/Shigella flexneri, caused a strong translocation, activation, and cytokines secretion but they could not inhibit TNF-α and EGF stimulation. ETEC and mainly EAEC caused a moderate translocation, but a differential activation, and high cytokines secretion; interestingly TNF-α and EGF stimulation did no modify p65 and ERK1/2 activation. The use of inhibitors of NF-κB and/or ERK1/2 showed that NF-κB is crucial for cytokine induction by the different pathotypes; only partially depended on ERK1/2 activation. Thus, in spite of their differences, the pathotypes can also be divided in three groups according to their inflammatory response as those (i) that inject effectors to cause A/E lesion, which are able to inhibit NF-κB and ERK1/2 pathways, and cytokine secretion; (ii) with fimbrial adherence and toxin secretion with a moderate inhibition of both pathways but high cytokines secretion through autocrine cytokine regulation; and (iii) the intracellular bacteria that induce the highest pathways activation and cytokines secretion by using different activation mechanisms. This study provides a comprehensive analysis of how the different pathogenesis schemes of E. coli pathotypes manipulate inflammatory signaling pathways, which leads to a specific proinflammatory cytokine secretion in a cell model infection that reproduce the hallmarks of infection of each pathotype.


Assuntos
Citocinas/metabolismo , Células Epiteliais/microbiologia , Escherichia coli/patogenicidade , Interações Hospedeiro-Patógeno , Transdução de Sinais , Western Blotting , Linhagem Celular , Humanos , Microscopia Confocal , Modelos Biológicos
4.
mBio ; 7(3)2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27329750

RESUMO

UNLABELLED: Enteropathogenic Escherichia coli (EPEC) has the ability to antagonize host apoptosis during infection through promotion and inhibition of effectors injected by the type III secretion system (T3SS), but the total number of these effectors and the overall functional relationships between these effectors during infection are poorly understood. EspC produced by EPEC cleaves fodrin, paxillin, and focal adhesion kinase (FAK), which are also cleaved by caspases and calpains during apoptosis. Here we show the role of EspC in cell death induced by EPEC. EspC is involved in EPEC-mediated cell death and induces both apoptosis and necrosis in epithelial cells. EspC induces apoptosis through the mitochondrial apoptotic pathway by provoking (i) a decrease in the expression levels of antiapoptotic protein Bcl-2, (ii) translocation of the proapoptotic protein Bax from cytosol to mitochondria, (iii) cytochrome c release from mitochondria to the cytoplasm, (iv) loss of mitochondrial membrane potential, (v) caspase-9 activation, (vi) cleavage of procaspase-3 and (vii) an increase in caspase-3 activity, (viii) PARP proteolysis, and (ix) nuclear fragmentation and an increase in the sub-G1 population. Interestingly, EspC-induced apoptosis was triggered through a dual mechanism involving both independent and dependent functions of its EspC serine protease motif, the direct cleavage of procaspase-3 being dependent on this motif. This is the first report showing a shortcut for induction of apoptosis by the catalytic activity of an EPEC protein. Furthermore, this atypical intrinsic apoptosis appeared to induce necrosis through the activation of calpain and through the increase of intracellular calcium induced by EspC. Our data indicate that EspC plays a relevant role in cell death induced by EPEC. IMPORTANCE: EspC, an autotransporter protein with serine protease activity, has cytotoxic effects on epithelial cells during EPEC infection. EspC causes cytotoxicity by cleaving fodrin, a cytoskeletal actin-associated protein, and focal adhesion proteins (i.e., FAK); interestingly, these proteins are also cleaved during apoptosis and necrosis. Here we show that EspC is able to cause cell death, which is characterized by apoptosis: by dissecting the apoptotic pathway and considering that EspC is translocated by an injectisome, we found that EspC induces the mitochondrial apoptotic pathway. Remarkably, EspC activates this pathway by two distinct mechanisms-either by using or not using its serine protease motif. Thus, we show for the first time that this serine protease motif is able to cleave procaspase-3, thereby reaching the terminal stages of caspase cascade activation leading to apoptosis. Furthermore, this overlapped apoptosis appears to potentiate cell death through necrosis, where EspC induces calpain activation and increases intracellular calcium.


Assuntos
Apoptose , Calpaína/metabolismo , Caspase 3/metabolismo , Caspases/metabolismo , Escherichia coli Enteropatogênica/patogenicidade , Proteínas de Escherichia coli/metabolismo , Necrose , Linhagem Celular , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Proteólise
5.
Infect Immun ; 84(7): 2012-2021, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27113356

RESUMO

Most autotransporter passenger domains, regardless of their diversity in function, fold or are predicted to fold as right-handed ß-helices carrying various loops that are presumed to confer functionality. Our goal here was to identify the subdomain (loop) or amino acid sequence of the Pet passenger domain involved in the receptor binding site on the host cell for Pet endocytosis. Here, we show that d1 and d2 subdomains, as well as the amino acid sequence linking the subdomain d2 and the adjacent ß-helix (PDWET), are not required for Pet secretion through the autotransporter system and that none of our deletion mutants altered the predicted long right-handed ß-helical structure. Interestingly, Pet lacking the d2 domain (PetΔd2) was unable to bind on the epithelial cell surface, in contrast to Pet lacking d1 (PetΔd1) subdomain or PDWET sequences. Moreover, the purified d1 subdomain, the biggest subdomain (29.8 kDa) containing the serine protease domain, was also unable to bind the cell surface. Thus, d2 sequence (54 residues without the PDWET sequence) was required for Pet binding to eukaryotic cells. In addition, this d2 sequence was also needed for Pet internalization but not for inducing cell damage. In contrast, PetΔd1, which was able to bind and internalize inside the cell, was unable to cause cell damage. Furthermore, unlike Pet, PetΔd2 was unable to bind cytokeratin 8, a Pet receptor. These data indicate that the surface d2 subdomain is essential for the ligand-receptor (Pet-Ck8) interaction for Pet uptake and to start the epithelial cell damage by this toxin.


Assuntos
Enterotoxinas/metabolismo , Células Epiteliais/metabolismo , Queratina-8/metabolismo , Domínios e Motivos de Interação entre Proteínas , Sistemas de Secreção Tipo V/metabolismo , Sítios de Ligação , Linhagem Celular , Membrana Celular/metabolismo , Enterotoxinas/química , Enterotoxinas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Humanos , Queratina-8/química , Ligantes , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Sistemas de Secreção Tipo V/genética
6.
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
7.
Infect Immun ; 82(6): 2255-65, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24643541

RESUMO

EspC is a non-locus of enterocyte effacement (LEE)-encoded autotransporter produced by enteropathogenic Escherichia coli (EPEC) that is secreted to the extracellular milieu by a type V secretion system and then translocated into epithelial cells by the type III secretion system. Here, we show that this efficient EspC delivery into the cell leads to a cytopathic effect characterized by cell rounding and cell detachment. Thus, EspC is the main protein involved in epithelial cell cytotoxicity detected during EPEC adhesion and pedestal formation assays. The cell detachment phenotype is triggered by cytoskeletal and focal adhesion disruption. EspC-producing EPEC is able to cleave fodrin, paxillin, and focal adhesion kinase (FAK), but these effects are not observed when cells are infected with an espC isogenic mutant. Recovery of these phenotypes by complementing the mutant with the espC gene but not with the espC gene mutated in the serine protease motif highlights the role of the protease activity of EspC in the cell detachment phenotype. In vitro assays using purified proteins showed that EspC, but not EspC with an S256I substitution [EspCS256I], directly cleaves these cytoskeletal and focal adhesion proteins. Kinetics of protein degradation indicated that EspC-producing EPEC first cleaves fodrin (within the 11th and 9th repetitive units at the Q1219 and D938 residues, respectively), and this event sequentially triggers paxillin degradation, FAK dephosphorylation, and FAK degradation. Thus, cytoskeletal and focal adhesion protein cleavage leads to the cell rounding and cell detachment promoted by EspC-producing EPEC.


Assuntos
Aderência Bacteriana/fisiologia , Proteínas de Transporte/metabolismo , Escherichia coli Enteropatogênica/patogenicidade , Células Epiteliais/metabolismo , Proteínas de Escherichia coli/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteínas dos Microfilamentos/metabolismo , Paxilina/metabolismo , Adesão Celular , Linhagem Celular , Células Epiteliais/patologia , Infecções por Escherichia coli/microbiologia , Humanos
8.
Infect Immun ; 82(4): 1719-24, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24516112

RESUMO

Fimbria-mediated adherence to the intestinal epithelia is a key step in enteroaggregative Escherichia coli (EAEC) pathogenesis. To date, four fimbriae have been described for EAEC; aggregative adherence fimbria II (AAF/II) is the most important adherence factor for EAEC prototype strain 042. Previously, we described results showing that extracellular matrix (ECM) components might be involved in the recognition of AAF/II fimbriae by intestinal cells. In this study, we sought to identify novel potential receptors on intestinal epithelial cells recognized by the AAF/II fimbriae. Purified AafA-dsc protein, the major subunit of AAF/II fimbriae, was incubated with a monolayer of T84 cells, cross-linked to the surface-exposed T84 cell proteins, and immunoprecipitated by using anti-AafA antibodies. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis of cellular proteins bound to AafA-dsc protein identified laminin (previously recognized as a potential receptor for AAF/II) and cytokeratin 8 (CK8). Involvement of the major subunit of AAF/II fimbriae (AafA protein) in the binding to recombinant CK8 was confirmed by adherence assays with purified AAF/II fimbriae, AafA-dsc protein, and strain 042. Moreover, HEp-2 cells transfected with CK8 small interfering RNA (siRNA) showed reduced 042 adherence compared with cells transfected with scrambled siRNA as a control. Adherence of 042 to HEp-2 cells preincubated with antibodies against ECM proteins or CK8 was substantially reduced. Altogether, our results supported the idea of a role of CK8 as a potential receptor for EAEC.


Assuntos
Aderência Bacteriana/fisiologia , Células Epiteliais/microbiologia , Escherichia coli/fisiologia , Fímbrias Bacterianas/fisiologia , Queratina-8/fisiologia , Laminina/fisiologia , Adesinas de Escherichia coli , Linhagem Celular , Células Epiteliais/fisiologia , Fibronectinas/imunologia , Humanos , Mucosa Intestinal/citologia , Queratina-8/metabolismo , Laminina/imunologia , Proteínas de Membrana
9.
mBio ; 4(6): e00838-13, 2013 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-24327340

RESUMO

UNLABELLED: The group of proteins known as serine protease autotransporters of Enterobacteriaceae (SPATE) is a growing family of serine proteases secreted to the external milieu by the type V secretion system. Pet toxin and some other SPATE belong to the class 1 cytotoxic SPATE, which have comparable protease strength on fodrin. Pet is internalized and is directed to its intracellular substrate by retrograde transport. However, the epithelial cell receptor for Pet has yet to be identified. We show that Pet has affinity for the epithelial cell surface until the saturation of the binding sites at 100 nM Pet. Affinity column assays and matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) analysis identified a cytokeratin (CK8) which directly binds to Pet, and both proteins colocalized on the cell surface. Interestingly, CK8 is not present in kidney cell lines, which are not susceptible to Pet. Inhibition experiments by using anti-CK8 and ck8 small interfering RNA (siRNA) blocked the cytotoxic effect induced by Pet, while exogenous CK8 expression in kidney cells made them susceptible to Pet intoxication. Recombinant CK8 showed a Pet-binding pattern similar to that seen by using fixed cells. Remarkably, Pet colocalized with CK8 and clathrin at early times (receptor-mediated endocytosis), and subsequently, Pet colocalized with CK8 and Rab5b in the early endosomes. These data support the idea that CK8 is an important receptor for Pet on epithelial cells for starting its cytotoxic effects. These data suggest that therapeutics that block Pet-CK8 interaction may improve outcome of diseases caused by Pet-secreting Enterobacteriaceae such as enteroaggregative Escherichia coli. IMPORTANCE: Receptor-ligand binding is one mechanism by which cells sense and respond to external cues. Receptors may also be utilized by toxins to mediate their own internalization. Pet toxin is secreted by enteroaggregative Escherichia coli, an organism that causes persistent diarrhea in children, traveler's diarrhea, and acute and persistent diarrhea in patients with HIV. Pet is a member of the family of serine protease autotransporters of Enterobacteriaceae (SPATE). SPATE in different pathogens are virulence factors, and Pet belongs to the class 1 cytotoxic SPATE, which have comparable protease strength on their biological substrate, fodrin (a cytoskeletal protein important for maintaining cell viability). To cleave fodrin, Pet enters the cells by clathrin-mediated endocytosis. This mechanism includes receptor-mediated endocytosis (a receptor-ligand complex triggers the endocytosis). We show that CK8 is an important receptor for Pet on epithelial cells and that it may be useful for identifying molecules that block the interaction of CK8 with Pet.


Assuntos
Toxinas Bacterianas/metabolismo , Enterotoxinas/metabolismo , Células Epiteliais/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Interações Hospedeiro-Patógeno , Queratina-8/metabolismo , Serina Endopeptidases/metabolismo , Animais , Linhagem Celular , Cães , Humanos , Ligação Proteica , Coelhos
10.
Biomed Res Int ; 2013: 374395, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23509714

RESUMO

The actin cytoskeleton is a dynamic structure necessary for cell and tissue organization, including the maintenance of epithelial barriers. Disruption of the epithelial barrier coincides with alterations of the actin cytoskeleton in several disease states. These disruptions primarily affect the paracellular space, which is normally regulated by tight junctions. Thereby, the actin cytoskeleton is a common and recurring target of bacterial virulence factors. In order to manipulate the actin cytoskeleton, bacteria secrete and inject toxins and effectors to hijack the host cell machinery, which interferes with host-cell pathways and with a number of actin binding proteins. An interesting model to study actin manipulation by bacterial effectors is Escherichia coli since due to its genome plasticity it has acquired diverse genetic mobile elements, which allow having different E. coli varieties in one bacterial species. These E. coli pathotypes, including intracellular and extracellular bacteria, interact with epithelial cells, and their interactions depend on a specific combination of virulence factors. In this paper we focus on E. coli effectors that mimic host cell proteins to manipulate the actin cytoskeleton. The study of bacterial effector-cytoskeleton interaction will contribute not only to the comprehension of the molecular causes of infectious diseases but also to increase our knowledge of cell biology.


Assuntos
Citoesqueleto de Actina/metabolismo , Escherichia coli/metabolismo , Escherichia coli Êntero-Hemorrágica/metabolismo , Escherichia coli Êntero-Hemorrágica/patogenicidade , Escherichia coli Enteropatogênica/metabolismo , Escherichia coli Enteropatogênica/patogenicidade , Células Epiteliais/microbiologia , Epitélio/metabolismo , Epitélio/microbiologia , Escherichia coli/patogenicidade , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/metabolismo , Humanos , Proteínas dos Microfilamentos/metabolismo , Virulência , Fatores de Virulência/metabolismo
11.
Ann Hepatol ; 11(1): 107-17, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22166569

RESUMO

Although Entamoeba dispar displays a similar morphology to Entamoeba histolytica, cellular and molecular studies have revealed significant differences between these two amoebae, including the former being characterized as non-pathogenic and the later as pathogenic. However, recent in vivo and in vitro experiments have shown that E. dispar strains of different origin are capable of causing liver damage and destroying cell culture lines in the presence of common intestinal bacteria. These results suggested that E. dispar may present pathogenic behavior according to the specific E. dispar strain, culture and environmental conditions. To investigate this possibility, we carried out in vivo and in vitro studies using a xenic strain E. dispar (ICB-ADO) isolated from a symptomatic non-dysenteric Brazilian patient. This strain was able to induce liver necrosis in a hamster model that was more severe than that produced by E. histolytica. The ICB-ADO isolate also caused significantly more destruction of cultured MDCK cells and increased loss of transepithelial resistance than did the E. histolytica. Xenic E. dispar exhibited high proteolytic activity, which was partially inhibited by the addition of cysteine-protease inhibitors. Based on our biochemical and molecular characterization of E. dispar (ICB-ADO) xenic culture and its ability to produce liver abscesses, we conclude that this specific strain can indeed produce tissue damage, distinct from the frequently used non- pathogenic E. dispar SAW 760 strain.


Assuntos
Entamoeba/classificação , Entamoeba/patogenicidade , Abscesso Hepático Amebiano/parasitologia , Fígado/parasitologia , Animais , Brasil , Células Cultivadas , Cricetinae , Modelos Animais de Doenças , Cães , Humanos , Técnicas In Vitro , Incidência , Rim/parasitologia , Rim/patologia , Fígado/patologia , Abscesso Hepático Amebiano/epidemiologia , Abscesso Hepático Amebiano/patologia , Masculino , Mesocricetus , Proteólise
12.
Gut Microbes ; 2(1): 13-24, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21637014

RESUMO

Enteroaggregative Escherichia coli (EAEC) is an emerging pathogen associated with acute and persistent diarrhea in children and adults. EAEC strains are very heterogeneous and the pathogenesis of EAEC diarrhea is complex and not completely understood. Studies have suggested three major features of EAEC pathogenesis: abundant adherence to the intestinal mucosa, elaboration of enterotoxins and cytotoxins, and induction of mucosal inflammation. Here, we discuss the role of the virulence factors involved in these three major features, focusing in the EAEC adhesion including fimbrial and afimbrial factors, EAEC toxins and autotransporter proteins, such as Pet (plasmid encoded toxin) and Pic (protein involved in colonization); both proteins play a role in two EAEC pathogenic features: cytotoxicity and mucosal colonization, including the bacterium-mucus biofilm. Finally we discuss relevant factors involved in the inflammatory process induce by EAEC, such as flagellin, fimbria and regulator factors (AggR). Interestingly, all these factors are not present in all EAEC strain, contributing to EAEC heterogeneity.


Assuntos
Proteínas de Transporte/metabolismo , Escherichia coli/patogenicidade , Serina Proteases/metabolismo , Fatores de Virulência/metabolismo , Aderência Bacteriana , Proteínas de Transporte/genética , Linhagem Celular , Enterotoxinas/genética , Enterotoxinas/metabolismo , Células Epiteliais/microbiologia , Escherichia coli/genética , Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Serina Proteases/genética , Virulência , Fatores de Virulência/genética
13.
J Biol Chem ; 285(41): 31261-7, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20667832

RESUMO

Cholera toxin (CT) is an AB(5) toxin that moves from the cell surface to the endoplasmic reticulum (ER) by retrograde vesicular transport. In the ER, the catalytic A1 subunit dissociates from the rest of the toxin and enters the cytosol by exploiting the quality control system of ER-associated degradation (ERAD). The driving force for CTA1 dislocation into the cytosol is unknown. Here, we demonstrate that the cytosolic chaperone Hsp90 is required for CTA1 passage into the cytosol. Hsp90 bound to CTA1 in an ATP-dependent manner that was blocked by geldanamycin (GA), an established Hsp90 inhibitor. CT activity against cultured cells and ileal loops was also blocked by GA, as was the ER-to-cytosol export of CTA1. Experiments using RNA interference or N-ethylcarboxamidoadenosine, a drug that inhibits ER-localized GRP94 but not cytosolic Hsp90, confirmed that the inhibitory effects of GA resulted specifically from the loss of Hsp90 activity. This work establishes a functional role for Hsp90 in the ERAD-mediated dislocation of CTA1.


Assuntos
Toxina da Cólera/metabolismo , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Trifosfato de Adenosina/genética , Trifosfato de Adenosina/metabolismo , Animais , Benzoquinonas/farmacologia , Células CHO , Toxina da Cólera/genética , Cricetinae , Cricetulus , Retículo Endoplasmático/genética , Inibidores Enzimáticos/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/genética , Células HeLa , Humanos , Lactamas Macrocíclicas/farmacologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ligação Proteica , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
14.
Future Microbiol ; 5(7): 1005-13, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20632801

RESUMO

Plasmid-encoded toxin (Pet) is secreted by enteroaggregative Escherichia coli (EAEC), a pathotype of diarrhogenic E. coli. EAEC infection is an important cause of diarrhea in outbreak and nonoutbreak settings in developing and developed countries. EAEC secretes Pet by using the type V secretion system. Mature secreted Pet is a serine protease and its eukaryotic target is the actin-binding protein alpha-fodrin. When Pet cleaves alpha-fodrin in the target cell cytosol, the organization of the actin cytoskeleton is disrupted. The loss of actin filament structure results in cell rounding and detachment from the substratum. This article summarizes the long trip of Pet during its biogenesis, its interaction with epithelial cells, intracellular trafficking and mechanism of action.


Assuntos
Toxinas Bacterianas/metabolismo , Enterotoxinas/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Serina Endopeptidases/metabolismo , Serina Proteases/metabolismo , Animais , Proteínas de Transporte/metabolismo , Citoesqueleto/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Proteínas dos Microfilamentos/metabolismo , Modelos Biológicos
15.
PLoS One ; 5(2): e9046, 2010 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-20140230

RESUMO

BACKGROUND: Enteropathogenic E. coli (EPEC) and enterohemorrhagic E. coli (EHEC) are two categories of E. coli strains associated with human disease. A major virulence factor of both pathotypes is the expression of a type three secretion system (TTSS), responsible for their ability to adhere to gut mucosa causing a characteristic attaching and effacing lesion (A/E). The TTSS translocates effector proteins directly into the host cell that subvert mammalian cell biochemistry. METHODS/PRINCIPAL FINDINGS: We examined synthetic peptides designed to inhibit the TTSS. CoilA and CoilB peptides, both representing coiled-coil regions of the translocator protein EspA, and CoilD peptide, corresponding to a coiled-coil region of the needle protein EscF, were effective in inhibiting the TTSS dependent hemolysis of red blood cells by the EPEC E2348/69 strain. CoilA and CoilB peptides also reduced the formation of actin pedestals by the same strain in HEp-2 cells and impaired the TTSS-mediated protein translocation into the epithelial cell. Interestingly, CoilA and CoilB were able to block EspA assembly, destabilizing the TTSS and thereby Tir translocation. This blockage of EspA polymerization by CoilA or CoilB peptides, also inhibited the correct delivery of EspB and EspD as detected by immunoblotting. Interestingly, electron microscopy of bacteria incubated with the CoilA peptide showed a reduction of the length of EspA filaments. CONCLUSIONS: Our data indicate that coiled-coil peptides can prevent the assembly and thus the functionality of the TTSS apparatus and suggest that these peptides could provide an attractive tool to block EPEC and EHEC pathogenesis.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Escherichia coli Enteropatogênica/efeitos dos fármacos , Escherichia coli Enteropatogênica/metabolismo , Proteínas de Escherichia coli/metabolismo , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Proteínas da Membrana Bacteriana Externa/metabolismo , Western Blotting , Linhagem Celular Tumoral , Proteínas do Citoesqueleto/química , Eritrócitos/efeitos dos fármacos , Eritrócitos/microbiologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/ultraestrutura , Hemólise/efeitos dos fármacos , Humanos , Viabilidade Microbiana/efeitos dos fármacos , Microscopia Imunoeletrônica , Dados de Sequência Molecular , Peptídeos/síntese química , Transporte Proteico/efeitos dos fármacos , Ovinos
16.
PLoS One ; 4(12): e8223, 2009 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-20011051

RESUMO

BACKGROUND: We have previously shown that the enterotoxin SigA which resides on the she pathogenicity island (PAI) of S. flexneri 2a is an autonomously secreted serine protease capable of degrading casein. We have also demonstrated that SigA is cytopathic for HEp-2 cells and plays a role in the intestinal fluid accumulation associated with S. flexneri infections. METHODS/PRINCIPAL FINDINGS: In this work we show that SigA binds specifically to HEp-2 cells and degrades recombinant human alphaII spectrin (alpha-fodrin) in vitro, suggesting that the cytotoxic and enterotoxic effects mediated by SigA are likely associated with the degradation of epithelial fodrin. Consistent with our data, this study also demonstrates that SigA cleaves intracellular fodrin in situ, causing its redistribution within cells. These results strongly implicate SigA in altering the cytoskeleton during the pathogenesis of shigellosis. On the basis of these findings, cleavage of fodrin is a novel mechanism of cellular intoxication for a Shigella toxin. Furthermore, information regarding immunogenicity to SigA in infected patients is lacking. We studied the immune response of SigA from day 28 post-challenge serum of one volunteer from S. flexneri 2a challenge studies. Our results demonstrate that SigA is immunogenic following infection with S. flexneri 2a. CONCLUSIONS: This work shows that SigA binds to epithelial HEp-2 cells as well as being able to induce fodrin degradation in vitro and in situ, further extending its documented role in the pathogenesis of Shigella infections.


Assuntos
Proteínas de Bactérias/imunologia , Proteínas de Transporte/metabolismo , Enterotoxinas/imunologia , Células Epiteliais/metabolismo , Proteínas dos Microfilamentos/metabolismo , Shigella flexneri/imunologia , Formação de Anticorpos/imunologia , Linhagem Celular , Humanos , Ligação Proteica , Transporte Proteico
17.
Microbiology (Reading) ; 155(Pt 10): 3214-3225, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19628559

RESUMO

Enterohaemorrhagic and enteropathogenic Escherichia coli (EHEC and EPEC) inject a repertoire of effector proteins into host cells via a type III secretion system (T3SS) encoded by the locus of enterocyte effacement (LEE). OspG is an effector protein initially identified in Shigella that was shown to inhibit the host innate immune response. In this study, we found ospG homologues in EHEC (mainly of serogroup O111) and in Yersinia enterocolitica. The T3SS encoded by the LEE was able to inject these different OspG homologues into host cells. Infection of HeLa cells with EHEC O111 inhibited the NF-kappaB-dependent innate immune response via a T3SS-dependent mechanism. However, an EHEC O111 ospG mutant was still able to inhibit NF-kappaB p65 transfer to the nucleus in infected cells stimulated by tumour necrosis factor alpha (TNF-alpha). In addition, no difference in the inflammatory response was observed between wild-type EHEC O111 and the isogenic ospG mutant in the rabbit ligated intestinal loop model. These results suggest that OspG is not the sole effector protein involved in the inactivation of the host innate immune system during EHEC O111 infection.


Assuntos
Escherichia coli Êntero-Hemorrágica/imunologia , Escherichia coli Êntero-Hemorrágica/patogenicidade , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/fisiologia , Imunidade Inata , NF-kappa B/antagonistas & inibidores , Fatores de Virulência/fisiologia , Sequência de Aminoácidos , Animais , Escherichia coli Enteropatogênica/imunologia , Escherichia coli Enteropatogênica/patogenicidade , Infecções por Escherichia coli/patologia , Ordem dos Genes , Células HeLa , Humanos , Intestinos/microbiologia , Intestinos/patologia , Dados de Sequência Molecular , Coelhos , Alinhamento de Sequência , Sorotipagem
18.
Microbiology (Reading) ; 155(Pt 9): 2895-2906, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19542001

RESUMO

In an in vitro model using HEp-2 cells treated with purified plasmid-encoded toxin (Pet), we have identified morphological changes characterized by cell rounding and detachment after toxin internalization; these changes progress to cell death. However, these effects have not yet been shown to occur during the infection of epithelial cells by enteroaggregative Escherichia coli (EAEC). Here, we show that the secretion of Pet by EAEC is regulated at the transcriptional level, since secretion was inhibited in eukaryotic cell culture medium, although Pet was efficiently secreted in the same medium supplemented with tryptone. Inefficient secretion of Pet by EAEC in DMEM prevented cell detachment, whereas efficient Pet secretion in DMEM/tryptone increased cell detachment in a HEp-2 cell adherence assay. Interestingly, Pet toxin was efficiently delivered to epithelial cells, since it was internalized into epithelial cells infected with EAEC at similar concentrations to those obtained by using 37 microg ml(-1) purified Pet protein. Additionally, Pet was not internalized when the epithelial cells were infected with a pet clone, HB101(pCEFN1), unlike the wild-type strain, which has a high adherence capability. There is a correlation between Pet secretion by EAEC, the internalization of Pet into epithelial cells, cell detachment and cell death in EAEC-infected cells. The ratio between live and dead cells decreased in cells treated with wild-type EAEC in comparison with cells treated with an isogenic mutant in the pet gene, whereas the effects were restored by complementing the mutant with the pet gene. All these data indicate that Pet is an important virulence factor in the pathogenesis of EAEC infection.


Assuntos
Toxinas Bacterianas , Morte Celular , Enterotoxinas , Células Epiteliais/fisiologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli , Escherichia coli/patogenicidade , Serina Endopeptidases , Toxinas Bacterianas/metabolismo , Adesão Celular , Linhagem Celular , Meios de Cultura , Enterotoxinas/metabolismo , Células Epiteliais/microbiologia , Escherichia coli/fisiologia , Infecções por Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Peptonas/metabolismo , RNA Bacteriano/biossíntese , RNA Bacteriano/genética , Serina Endopeptidases/metabolismo , Ativação Transcricional , Regulação para Cima , Virulência , Fatores de Virulência/metabolismo
19.
Can J Microbiol ; 54(9): 748-57, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18772938

RESUMO

Enteropathogenic Escherichia coli (EPEC) colonizes the intestinal mucosa and causes a cell lesion known as attachment and effacement (A/E) lesion. The molecular mechanisms for A/E lesions include injection of Tir, which is a receptor for an adhesin named intimin. The Tir-intimin interaction causes rearrangement of the cytoskeleton forming actin-rich structures called pedestals. Unfortunately, the formation of the A/E lesions and the dynamics of the actin cytoskeleton during this rearrangement induced by EPEC cannot be studied in the natural host. However, there are EPEC strains that infect rabbit (REPEC) that are genetically and pathologically similar to EPEC. Here, we used REPEC for the infection of rabbit kidney epithelial cells, line RK13, as a model to understand the actin cytoskeleton dynamics during pedestal formation. Actin-rich pedestal formation during the infection of RK13 cells by REPEC was analyzed by electron and confocal microscopy. The kinetics of infection along with the use of antibiotics for eliminating the bacteria, as well as reinfection, evidenced the plasticity of the actin cytoskeleton during pedestal formation. Thus, this model is a helpful tool for studying the dynamics of actin cytoskeleton and for correlating the data with those observed in in vivo models in rabbits experimentally infected with REPEC.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Escherichia coli Enteropatogênica/fisiologia , Células Epiteliais/metabolismo , Infecções por Escherichia coli/metabolismo , Modelos Biológicos , Actinas/química , Actinas/ultraestrutura , Animais , Linhagem Celular , Citoesqueleto/química , Citoesqueleto/microbiologia , Citoesqueleto/ultraestrutura , Escherichia coli Enteropatogênica/patogenicidade , Células Epiteliais/química , Células Epiteliais/microbiologia , Células Epiteliais/ultraestrutura , Infecções por Escherichia coli/microbiologia , Humanos , Coelhos
20.
Cell Microbiol ; 10(10): 1975-86, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18547338

RESUMO

EspC is a non-locus of enterocyte effacement (LEE)-encoded autotransporter protein secreted by enteropathogenic Escherichia coli (EPEC) that causes a cytopathic effect on epithelial cells, including cytoskeletal damage. EspC cytotoxicity depends on its internalization and functional serine protease motif. Here we show that during EPEC infection, EspC is secreted from the bacteria by the type V secretion system (T5SS) and then it is efficiently translocated into the epithelial cells through the type III secretion system (T3SS) translocon. By dissecting this mechanism, we found that EspC internalization during EPEC-host cell interaction occurs after 1 h, unlike purified EspC (8 h). LEE pathogenicity island is involved in specific EspC translocation as three espC-transformed attaching and effacing (AE) pathogens translocated EspC into the cells. A role for effectors and other factors involved in the intimate adherence encoded in LEE were discarded by using an exogenous EspC internalization model. In this model, an isogenic EPEC DeltaespC strain allows the efficient internalization of purified EspC. Moreover, isogenic mutants in T3SS were unable to translocate endogenous and exogenous EspC into epithelial cells, as EspC-EspA interaction is required. These data show for the first time the efficient delivery of an autotransporter protein inside the epithelial cells by EPEC, through cooperation between T5SS and T3SS.


Assuntos
Escherichia coli Enteropatogênica/metabolismo , Células Epiteliais/microbiologia , Proteínas de Escherichia coli/metabolismo , Linhagem Celular , Escherichia coli Enteropatogênica/genética , Proteínas de Escherichia coli/genética , Deleção de Genes , Ilhas Genômicas , Humanos , Imunoprecipitação , Microscopia Confocal , Fosfoproteínas/metabolismo , Ligação Proteica , Transporte Proteico , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA