Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Gut Microbes ; 12(1): 1-21, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33131419

RESUMO

Infectious diarrhea causes approximately 179 million illnesses annually in the US. Multiplex PCR assays for enteric pathogens detect enteropathogenic Escherichia coli (EPEC) in 12-29% of diarrheal stool samples from all age groups in developed nations. The aim of this study was to isolate and characterize EPEC from diarrhea samples identified as EPEC positive by BioFire Gastrointestinal Panel (GIP). EPEC is the second most common GIP-detected pathogen, equally present in sole and mixed infections peaking during summer months. EPEC bacterial load is higher in samples with additional pathogens. EPEC-GIP-positive stool samples were cultured on MacConkey II agar and analyzed by colony PCR for eaeA and bfpA to identify and classify EPEC isolates as typical (tEPEC) or atypical (aEPEC). EPEC were not recovered from the majority of stool samples with only 61 isolates obtained from 277 samples; most were aEPEC from adults. bfpA-mRNA was severely diminished in 3 of 4 bfpA-positive isolates. HeLa and SKCO-15 epithelial cells were infected with EPEC isolates and virulence-associated phenotypes, including adherence pattern, attachment level, pedestal formation, and tight junction disruption, were assessed. All aEPEC adherence patterns were represented with diffuse adherence predominating. Attachment rates of isolates adhering with defined adherence patterns were higher than tEPEC lacking bfpA (ΔbfpA). The majority of isolates formpedestals. All but one isolate initially increases but ultimately decreases transepithelial electrical resistance of SKCO-15 monolayers, similar to ΔbfpA. Most isolates severely disrupt occludin; ZO-1 disruption is variable. Most aEPEC isolates induce more robust virulence-phenotypes in vitro than ΔbfpA, but less than tEPEC-E2348/69.


Assuntos
Escherichia coli Enteropatogênica/genética , Escherichia coli Enteropatogênica/patogenicidade , Gastroenterite/microbiologia , Fatores de Virulência/genética , Adesinas Bacterianas/genética , Adulto , Aderência Bacteriana/fisiologia , Carga Bacteriana , Linhagem Celular Tumoral , Diarreia/microbiologia , Escherichia coli Enteropatogênica/isolamento & purificação , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Feminino , Proteínas de Fímbrias/genética , Fímbrias Bacterianas/genética , Genoma Bacteriano/genética , Células HeLa , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Adulto Jovem
2.
Gastroenterology ; 157(6): 1544-1555.e3, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31473225

RESUMO

BACKGROUND & AIMS: Sperm flagellar 1 (also called CLAMP) is a microtubule-associated protein that regulates microtubule dynamics and planar cell polarity in multi-ciliated cells. We investigated the localization and function of sperm flagellar 1, or CLAMP, in human intestinal epithelia cells (IECs). METHODS: We performed studies with SKCO-15 and human intestinal enteroids established from biopsies from different intestinal segments (duodenal, jejunum, ileal, and colon) of a single donor. Enteroids were induced to differentiation after incubation with growth factors. The distribution of endogenous CLAMP in IECs was analyzed by immunofluorescence microscopy using total internal reflection fluorescence-ground state depletion and confocal microscopy. CLAMP localization was followed during the course of intestinal epithelial cell polarization as cells progressed from flat to compact, confluent monolayers. Protein interactions with endogenous CLAMP were determined in SKCO-15 cells using proximity ligation assays and co-immunoprecipitation. CLAMP was knocked down in SKCO-15 monolayers using small hairpin RNAs and cells were analyzed by immunoblot and immunofluorescence microscopy. The impact of CLAMP knock-down in migrating SKCO-15 cells was assessed using scratch-wound assays. RESULTS: CLAMP bound to actin and apical junctional complex proteins but not microtubules in IECs. In silico analysis predicted the calponin-homology domain of CLAMP to contain conserved amino acids required for actin binding. During IEC polarization, CLAMP distribution changed from primarily basal stress fibers and cytoplasm in undifferentiated cells to apical membranes and microvilli in differentiated monolayers. CLAMP accumulated in lamellipodia and filopodia at the leading edge of migrating cells in association with actin. CLAMP knock-down reduced the number of filopodia, perturbed filopodia polarity, and altered the organization of actin filaments within lamellipodia. CONCLUSIONS: CLAMP is an actin-binding protein, rather than a microtubule-binding protein, in IECs. CLAMP distribution changes during intestinal epithelial cell polarization, regulates the formation of filopodia, and appears to assist in the organization of actin bundles within lamellipodia of migrating IECs. Studies are needed to define the CLAMP domains that interact with actin and whether its loss from IECs affects intestinal function.


Assuntos
Actinas/metabolismo , Movimento Celular , Mucosa Intestinal/citologia , Proteínas dos Microfilamentos/metabolismo , Pseudópodes/metabolismo , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Colo/citologia , Colo/metabolismo , Células Epiteliais , Humanos , Mucosa Intestinal/metabolismo , Microtúbulos/metabolismo
3.
Ann N Y Acad Sci ; 1405(1): 16-24, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28628193

RESUMO

Epithelial cells constitute a physical barrier that aids in protecting the host from microbial pathogens. Polarized epithelial cells contain distinct apical and basolateral membrane domains separated by intercellular junctions, including tight junctions (TJs), which contribute to the maintenance of apical-basal polarity. Polarity complexes also contribute to the establishment of TJ formation. Several pathogens perturb epithelial TJ barrier function and structure in addition to causing a loss of apical-basal polarity. Here, we review the impact of pathogenic bacteria on the disruption of cell-cell junctions and epithelial polarity.


Assuntos
Polaridade Celular/fisiologia , Células Epiteliais/citologia , Escherichia coli , Proteínas de Membrana/metabolismo , Junções Íntimas/microbiologia , Animais , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Humanos , Junções Íntimas/metabolismo
4.
Cell Microbiol ; 19(11)2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28618099

RESUMO

Enteropathogenic Escherichia coli (EPEC) uses a type III secretion system to inject effector proteins into host intestinal epithelial cells causing diarrhoea. EPEC infection redistributes basolateral proteins ß1-integrin and Na+ /K+ ATPase to the apical membrane of host cells. The Crumbs (Crb) polarity complex (Crb3/Pals1/Patj) is essential for epithelial cell polarisation and tight junction (TJ) assembly. Here, we demonstrate that EPEC displaces Crb3 and Pals1 from the apical membrane to the cytoplasm of cultured intestinal epithelial cells and colonocytes of infected mice. In vitro studies show that EspF, but not Map, alters Crb3, whereas both effectors modulate Pals1. EspF perturbs polarity formation in cyst morphogenesis assays and induces endocytosis and apical redistribution of Na+ /K+ ATPase. EspF binds to sorting nexin 9 (SNX9) causing membrane remodelling in host cells. Infection with ΔespF/pespFD3, a mutant strain that ablates EspF binding to SNX9, or inhibition of dynamin, attenuates Crb3 endocytosis caused by EPEC. In addition, infection with ΔespF/pespFD3 has no impact on Na+ /K+ ATPase endocytosis. These data support the hypothesis that EPEC perturbs apical-basal polarity in an EspF-dependent manner, which would contribute to EPEC-associated diarrhoea by disruption of TJ and altering the crucial positioning of membrane transporters involved in the absorption of ions and solutes.


Assuntos
Proteínas de Transporte/metabolismo , Escherichia coli Enteropatogênica/patogenicidade , Infecções por Escherichia coli/patologia , Proteínas de Escherichia coli/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Núcleosídeo-Fosfato Quinase/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Nexinas de Classificação/metabolismo , Animais , Proteínas de Transporte/genética , Linhagem Celular , Membrana Celular/metabolismo , Polaridade Celular/fisiologia , Antiportadores de Cloreto-Bicarbonato/antagonistas & inibidores , Diarreia/microbiologia , Diarreia/patologia , Cães , Dinaminas/antagonistas & inibidores , Endocitose/fisiologia , Células Epiteliais/fisiologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Humanos , Mucosa Intestinal/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Células Madin Darby de Rim Canino , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transportadores de Sulfato , Junções Íntimas/metabolismo , Sistemas de Secreção Tipo III/metabolismo
5.
Am J Physiol Gastrointest Liver Physiol ; 312(5): G443-G449, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28209599

RESUMO

Enteropathogenic Escherichia coli (EPEC), one of the diarrheagenic E. coli pathotypes, is among the most important food-borne pathogens infecting children worldwide. Inhibition of serotonin transporter (SERT), which regulates extracellular availability of serotonin (5-HT), has been implicated previously in EPEC-associated diarrhea. EPEC was shown to inhibit SERT via activation of protein tyrosine phosphatase (PTPase), albeit the specific PTPase involved is not known. Current studies aimed to identify EPEC-activated PTPase and its role in SERT inhibition. Infection of Caco-2 monolayers with EPEC strain E2348/69 for 30 min increased the activity of Src-homology-2 domain containing PTPase (SHP2) but not SHP1 or PTPase 1B. Similarly, Western blot studies showed increased tyrosine phosphorylation of (p-tyrosine) SHP2, indicative of its activation. Concomitantly, EPEC infection decreased SERT p-tyrosine levels. This was associated with increased interaction of SHP2 with SERT, as evidenced by coimmunoprecipitation studies. To examine whether SHP2 directly influences SERT phosphorylation status or function, SHP2 cDNA plasmid constructs (wild type, constitutively active, or dominant negative) were overexpressed in Caco-2 cells by Amaxa electroporation. In the cells overexpressing constitutively active SHP2, SERT polypeptide showed complete loss of p-tyrosine. In addition, there was a decrease in SERT function, as measured by Na+Cl--sensitive [3H]5-HT uptake, and an increase in association of SERT with SHP2 in Caco-2 cells expressing constitutively active SHP2 compared with dominant-negative SHP2. Our data demonstrate that intestinal SERT is a target of SHP2 and reveal a novel mechanism by which a common food-borne pathogen uses cellular SHP2 to inhibit SERT.NEW & NOTEWORTHY The data presented in the current study reveal that intestinal serotonin transporter (SERT) is a target of the tyrosine phosphatase SHP2 and show a novel mechanism by which a common diarrheagenic pathogen, EPEC, activates cellular SHP2 to inhibit SERT function. These studies highlight host-pathogen interactions, which may be of therapeutic relevance in the management of diarrhea associated with enteric infections.


Assuntos
Enterócitos/metabolismo , Enterócitos/microbiologia , Escherichia coli Enteropatogênica/metabolismo , Escherichia coli/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Células CACO-2 , Humanos
6.
Am J Physiol Cell Physiol ; 309(12): C835-46, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26447204

RESUMO

Enteropathogenic Escherichia coli (EPEC) is a food-borne pathogen that causes infantile diarrhea worldwide. EPEC decreases the activity and surface expression of the key intestinal Cl(-)/HCO3(-) exchanger SLC26A3 [downregulated in adenoma (DRA)], contributing to the pathophysiology of early diarrhea. Little is known about the mechanisms governing membrane recycling of DRA. In the current study, Caco-2 cells were used to investigate DRA trafficking under basal conditions and in response to EPEC. Apical Cl(-)/HCO3(-) exchange activity was measured as DIDS-sensitive (125)I(-) uptake. Cell surface biotinylation was performed to assess DRA endocytosis and exocytosis. Inhibition of clathrin-mediated endocytosis by chlorpromazine (60 µM) increased apical Cl(-)/HCO3(-) exchange activity. Dynasore, a dynamin inhibitor, also increased function and surface levels of DRA via decreased endocytosis. Perturbation of microtubules by nocodazole revealed that intact microtubules are essential for basal exocytic (but not endocytic) DRA recycling. Mice treated with colchicine showed a decrease in DRA surface levels as visualized by confocal microscopy. In response to EPEC infection, DRA surface expression was reduced partly via an increase in DRA endocytosis and a decrease in exocytosis. These effects were dependent on the EPEC virulence genes espG1 and espG2. Intriguingly, the EPEC-induced decrease in DRA function was unaltered in the presence of dynasore, suggesting a clathrin-independent internalization of surface DRA. In conclusion, these studies establish the role of clathrin-mediated endocytosis and microtubules in the basal surface expression of DRA and demonstrate that the EPEC-mediated decrease in DRA function and apical expression in Caco-2 cells involves decreased exocytosis.


Assuntos
Antiporters/metabolismo , Antiportadores de Cloreto-Bicarbonato/metabolismo , Escherichia coli Enteropatogênica , Infecções por Escherichia coli/metabolismo , Mucosa Intestinal/metabolismo , Animais , Western Blotting , Células CACO-2 , Clatrina/metabolismo , Endocitose/fisiologia , Exocitose/fisiologia , Imunofluorescência , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Transportadores de Sulfato
7.
Infect Immun ; 82(9): 3713-22, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24958711

RESUMO

Attachment of enterohemorrhagic Escherichia coli (EHEC) to intestinal epithelial cells is critical for colonization and is associated with localized actin assembly beneath bound bacteria. The formation of these actin "pedestals" is dependent on the translocation of effectors into mammalian cells via a type III secretion system (T3SS). Tir, an effector required for pedestal formation, localizes in the host cell plasma membrane and promotes attachment of bacteria to mammalian cells by binding to the EHEC outer surface protein Intimin. Actin pedestal formation has been shown to foster intestinal colonization by EHEC in some animal models, but the mechanisms responsible for this remain undefined. Investigation of the role of Tir-mediated actin assembly promoting host cell binding is complicated by other, potentially redundant EHEC-encoded binding pathways, so we utilized cell binding assays that specifically detect binding mediated by Tir-Intimin interaction. We also assessed the role of Tir-mediated actin assembly in two-step assays that temporally segregated initial translocation of Tir from subsequent Tir-Intimin interaction, thereby permitting the distinction of effects on translocation from effects on cell attachment. In these experimental systems, we compromised Tir-mediated actin assembly by chemically inhibiting actin assembly or by infecting mammalian cells with EHEC mutants that translocate Tir but are specifically defective in Tir-mediated pedestal formation. We found that an inability of Tir to promote actin assembly resulted in a significant and striking decrease in bacterial binding mediated by Tir and Intimin. Bacterial mutants defective for pedestal formation translocated type III effectors to mammalian cells with reduced efficiency, but the decrease in translocation could be entirely accounted for by the decrease in host cell attachment.


Assuntos
Actinas/metabolismo , Aderência Bacteriana/fisiologia , Translocação Bacteriana/fisiologia , Escherichia coli Êntero-Hemorrágica/metabolismo , Proteínas de Escherichia coli/metabolismo , Adesinas Bacterianas/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Ligação Proteica/fisiologia
8.
Am J Physiol Cell Physiol ; 307(3): C245-54, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24920678

RESUMO

Epithelial tight junctions are critical for creating a barrier yet allowing paracellular transport. Although it is well established that the actin cytoskeleton is critical for preserving the dynamic organization of the tight junction and maintaining normal tight junction protein recycling, contributions of microtubules to tight junction organization and function remain undefined. The aim of this study is to determine the role of microtubules in tight junction homeostasis and restoration. Our data demonstrate that occludin traffics on microtubules and that microtubule disruption perturbs tight junction structure and function. Microtubules are also shown to be required for restoring barrier function following Ca(2+) chelation and repletion. These processes are mediated by proteins participating in microtubule minus-end-directed trafficking but not plus-end-directed trafficking. These studies show that microtubules participate in the preservation of epithelial tight junction structure and function and play a vital role in tight junction restoration, thus expanding our understanding of the regulation of tight junction physiology.


Assuntos
Epitélio/metabolismo , Microtúbulos/metabolismo , Ocludina/metabolismo , Junções Íntimas/metabolismo , Citoesqueleto de Actina/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Compostos de Benzil/farmacologia , Células CACO-2 , Cálcio/química , Linhagem Celular Tumoral , Cães , Complexo Dinactina , Dineínas/antagonistas & inibidores , Dineínas/genética , Dineínas/metabolismo , Células Epiteliais/metabolismo , Complexo de Golgi/genética , Homeostase , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Células Madin Darby de Rim Canino , Proteínas Associadas aos Microtúbulos/genética , Nocodazol/farmacologia , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno , Moduladores de Tubulina/farmacologia
9.
Am J Physiol Gastrointest Liver Physiol ; 297(4): G825-33, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19628653

RESUMO

Infection with the gram-negative enteropathogenic Escherichia coli (EPEC), a food-borne pathogen, represents a significant risk to human health. Whereas diarrhea is a major consequence of this infection, malnutrition also occurs especially in severe and prolonged cases, which may aggravate the health status of the infected hosts. Here we examined the effect of EPEC infection on the intestinal uptake of the water-soluble vitamin B1 (thiamin) using an established human intestinal epithelial Caco-2 cell model. The results showed that infecting Caco-2 cells with wild-type EPEC (but not with nonpathogenic E. coli, killed EPEC, or filtered supernatant) leads to a significant (P < 0.01) inhibition in thiamin uptake. Kinetic parameters of both the nanomolar (mediated by THTR-2) and the micromolar (mediated by THTR-1) saturable thiamin uptake processes were affected by EPEC infection. Cell surface expression of hTHTR-1 and -2 proteins, (determined by the biotinylation method) showed a significantly (P < 0.01) lower expression in EPEC-treated cells compared with controls. EPEC infection also affected the steady-state mRNA levels as well as promoter activity of the SLC19A2 and SLC19A3 genes. Infecting Caco-2 cells with EPEC mutants that harbor mutations in the escN gene (which encodes a putative ATPase for the EPEC type III secretion system, TTSS) or the espA, espB, or espD genes (which encode structural components of the TTSS) did not affect thiamin uptake. On the other hand, mutations in espF and espH genes (which encode effector proteins) exhibited partial inhibition in thiamin uptake. These results demonstrate for the first time that EPEC infection of human intestinal epithelial cells leads to inhibition in thiamin uptake via effects on physiological and molecular parameters of hTHTR-1 and -2. Furthermore, the inhibition appears to be dependent on a functional TTSS of EPEC.


Assuntos
Células Epiteliais/microbiologia , Escherichia coli/patogenicidade , Mucosa Intestinal/microbiologia , Tiamina/metabolismo , Transporte Biológico , Células CACO-2 , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Regulação para Baixo , Células Epiteliais/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Cinética , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Viabilidade Microbiana , Mutação , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Transfecção
10.
J Nutr ; 138(7): 1355-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18567760

RESUMO

Probiotics are viable nonpathogenic microorganisms that are considered to confer health benefits to the host. Recent studies indicated that some Lactobacillus species function as probiotics and have been used as alternative treatments for diarrhea, which occurs due to increased secretion, decreased absorption, or both. However, the direct effects of probiotics on intestinal electrolyte absorption are not known. Therefore, we examined the effects of Lactobacillus on luminal chloride/hydroxyl (Cl(-)/OH(-)) exchange activity in human intestinal epithelial cells. Postconfluent Caco-2 cells were treated with the Lactobacillus species Lactobacillus acidophilus (LA), Lactobacillus casei, Lactobacillus plantarum, or Lactobacillus rhamnosus (LR) for 3 h at a multiplicity of infection of 50. Cl(-)/OH(-) exchange activity was measured as 4,4'-diisothiocyanostilbene-2, 2'-disulfonic acid-sensitive (36)Cl uptake in base-loaded cells. Treatment with live, but not heat-killed, LA and LR significantly increased Cl(-)/OH(-) exchange activity (approximately 50%), whereas other species were ineffective. Similarly, the conditioned medium (supernatant) of live LA increased Cl(-)/OH(-) exchange. The ability of LA or its conditioned culture medium to enhance Cl(-)/OH(-) exchange activity was blocked by PI-3 kinase inhibition but was unaffected by inhibition of mitogen-activated protein kinases. Corresponding to the increased Cl(-)/OH(-) exchange activity, LA treatment increased the surface expression of the apical anion exchanger, SLC26A3 [Down Regulated in Adenoma (DRA)]. The increased DRA membrane localization might contribute to the increased Cl(-) absorption by LA. Our results suggest that LA secretes soluble effector molecule(s) into the culture medium that stimulate apical Cl(-)/OH(-) exchange activity via phosphatidylinositol-3 kinase mediated mechanism.


Assuntos
Antiporters/metabolismo , Cloretos/metabolismo , Mucosa Intestinal/metabolismo , Lactobacillus acidophilus , Probióticos , Células CACO-2 , Antiportadores de Cloreto-Bicarbonato , Células Epiteliais/metabolismo , Humanos , Hidróxidos/metabolismo , Absorção Intestinal , Troca Iônica , Lacticaseibacillus casei , Lactobacillus plantarum , Lacticaseibacillus rhamnosus , Fosfatidilinositol 3-Quinases/metabolismo , Transportadores de Sulfato
12.
J Cell Biol ; 178(7): 1265-78, 2007 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-17893247

RESUMO

Bacterial toxins and effector proteins hijack eukaryotic enzymes that are spatially localized and display rapid signaling kinetics. However, the molecular mechanisms by which virulence factors engage highly dynamic substrates in the host cell environment are poorly understood. Here, we demonstrate that the enteropathogenic Escherichia coli (EPEC) type III effector protein EspF nucleates a multiprotein signaling complex composed of eukaryotic sorting nexin 9 (SNX9) and neuronal Wiskott-Aldrich syndrome protein (N-WASP). We demonstrate that a specific and high affinity association between EspF and SNX9 induces membrane remodeling in host cells. These membrane-remodeling events are directly coupled to N-WASP/Arp2/3-mediated actin nucleation. In addition to providing a biochemical mechanism of EspF function, we find that EspF dynamically localizes to membrane-trafficking organelles in a spatiotemporal pattern that correlates with SNX9 and N-WASP activity in living cells. Thus, our findings suggest that the EspF-dependent assembly of SNX9 and N-WASP represents a novel form of signaling mimicry used to promote EPEC pathogenesis and gastrointestinal disease.


Assuntos
Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Células Eucarióticas/metabolismo , Transdução de Sinais , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Actinas/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Proteínas de Transporte/química , Polaridade Celular , Sobrevivência Celular , Cães , Células Epiteliais/citologia , Proteínas de Escherichia coli/química , Evolução Molecular , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Ligantes , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Transporte Proteico , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/metabolismo , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo , Domínios de Homologia de src
13.
Lab Invest ; 85(10): 1308-24, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16127426

RESUMO

Enteropathogenic Escherichia coli (EPEC) infection disrupts tight junctions (TJs) and perturbs intestinal barrier function in vitro. E. coli secreted protein F (EspF) is, in large part, responsible for these physiological and morphological alterations. We recently reported that the C57BL/6J mouse is a valid in vivo model of EPEC infection as EPEC colonizes the intestinal epithelium and effaces microvilli. Our current aim was to examine the effects of EPEC on TJ structure and barrier function of the mouse intestine and to determine the role of EspF in vivo. C57BL/6J mice were gavaged with approximately 2 x 10(8) EPEC organisms or PBS. At 1 or 5 days postinfection, mice were killed and ileal and colonic tissue was mounted in Ussing chambers to determine barrier function (measured as transepithelial resistance) and short circuit current. TJ structure was analyzed by immunofluorescence microscopy. Wild-type (WT) EPEC significantly diminished the barrier function of ileal and colonic mucosa at 1 and 5 days postinfection. Deficits in barrier function correlated with redistribution of occludin in both tissues. Infection with an EPEC strain deficient of EspF (delta espF) had no effect on barrier function at 1 day postinfection. Furthermore, delta espF had no effect on ileal TJ morphology and minor alterations of colonic TJ morphology at 1 day postinfection. In contrast, at 5 days postinfection, WT EPEC and delta espF had similar effects on barrier function and occludin localization. In both cases this was associated with immune activation, as demonstrated by increased mucosal tumor necrosis factor-alpha levels 5 days postinfection. In conclusion, these data demonstrate that WT EPEC infection of 6-8-week-old C57BL/6J mice (1) significantly decreases barrier function in the ileum and colon (2) redistributes occludin in the ileum and colon and (3) is dependent upon EspF to induce TJ barrier defects at early, but not late, times postinfection.


Assuntos
Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/fisiologia , Escherichia coli/patogenicidade , Mucosa Intestinal/patologia , Proteínas de Membrana/metabolismo , Junções Íntimas/microbiologia , Animais , Colo/microbiologia , Colo/patologia , Colo/ultraestrutura , Enterócitos/metabolismo , Enterócitos/patologia , Enterócitos/ultraestrutura , Escherichia coli/genética , Escherichia coli/metabolismo , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/patologia , Proteínas de Escherichia coli/genética , Íleo/microbiologia , Íleo/patologia , Íleo/ultraestrutura , Mucosa Intestinal/microbiologia , Mucosa Intestinal/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Ocludina , Permeabilidade , Junções Íntimas/patologia , Junções Íntimas/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
14.
Cell Microbiol ; 6(8): 783-93, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15236645

RESUMO

Infection of intestinal epithelial cells with enteropathogenic Escherichia coli (EPEC) disrupts tight junction (TJ) architecture and barrier function. The aim of this study was to determine the impact of EPEC on TJ protein interactions and localization. Human intestinal epithelial cells (T84) were infected for 1, 3 or 6 h with EPEC. To probe the TJ protein-protein interactions, co-immunoprecipitations were performed. The associations between ZO-1, occludin and claudin-1 progressively decreased after infection. Corresponding morphological changes were analysed by immunofluorescence confocal microscopy. Tight junction proteins progressively lost their apically restricted localization. Freeze-fracture electron microscopy revealed the appearance of aberrant strands throughout the lateral membrane that contained claudin-1 and occludin as determined by immunogold labelling. These structural alterations were accompanied by a loss of barrier function. Mutation of the gene encoding EspF, important in the disruption of TJs by EPEC, prevented the disruption of TJs. Tight junction structure normalized following eradication of EPEC with gentamicin and overnight recovery. This is the first demonstration that a microbial pathogen can cause aberrant TJ strands in the lateral membrane of host cells. We speculate that the disruption of integral and cytoplasmic TJ protein interactions following EPEC infection allows TJ strands to form or diffuse into the lateral plasma membrane.


Assuntos
Células Epiteliais/microbiologia , Escherichia coli/patogenicidade , Intestino Delgado/microbiologia , Junções Íntimas/microbiologia , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Membrana Celular/ultraestrutura , Claudina-1 , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Técnica de Fratura por Congelamento , Humanos , Intestino Delgado/metabolismo , Intestino Delgado/ultraestrutura , Proteínas de Membrana/metabolismo , Microscopia Eletrônica , Ocludina , Fosfoproteínas/metabolismo , Mapeamento de Interação de Proteínas , Junções Íntimas/ultraestrutura , Proteína da Zônula de Oclusão-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA