Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Psychiatry ; 27(3): 1640-1646, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34969978

RESUMO

In contrast to healthy controls, the heterotrimeric G protein, Gsalpha (Gsα) is ensconced predominantly in lipid rafts in subjects with major depressive disorder (MDD) resulting in impaired stimulation of adenylyl cyclase. In this small proof-of-concept study, we examined the hypothesis that translocation of Gsα from lipid rafts toward a more facile activation of adenylyl cyclase is a biomarker for clinical response to antidepressants. There were 49 subjects with MDD (HamD17 score ≥15) and 59 healthy controls at the screen visit. The AlphaScreen (PerkinElmer) assay measured both basal activity and prostaglandin E1 (PGE1) stimulation of Gsα-adenylyl cyclase to assess the extent of coupling of Gsα with adenylyl cyclase. At screen, platelet samples obtained from MDD subjects revealed significantly lower PGE1 activation of adenylyl cyclase activity than controls (p = 0.02). Subsequently, 19 consenting MDD subjects completed a 6-week open label antidepressant treatment trial. The 11 antidepressant responders (HamD17 improvement ≥50% from screen) revealed significant increase in PGE1-stimulated adenylyl cyclase compared to non-responders (p = 0.05) with an effect size of 0.83 for the PGE1/Gsα lipid-raft biomarker. PGE1 stimulation increased by ≥30% from screen assessment in eight responders (72.7%) and two non-responders (25.0%) [Fisher exact = 0.07] with a positive predictive value for response of 80.0%. In this small, pilot study, increased PGE1 stimulated adenylyl cyclase was associated with antidepressant response in MDD subjects. These data suggest that a simple, high-throughput-capable assay for depression and antidepressant response can be developed. Future studies are needed to evaluate the utility of this biomarker for the treatment of MDD.


Assuntos
Adenilil Ciclases , Transtorno Depressivo Maior , Adenilil Ciclases/metabolismo , Alprostadil , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Biomarcadores , Depressão/tratamento farmacológico , Transtorno Depressivo Maior/tratamento farmacológico , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Humanos , Projetos Piloto
2.
J Biol Chem ; 290(16): 10045-56, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25691569

RESUMO

Signals that activate the G protein Gαs and promote neuronal differentiation evoke Gαs internalization in rat pheochromocytoma (PC12) cells. These agents also significantly increase Gαs association with microtubules, resulting in an increase in microtubule dynamics because of the activation of tubulin GTPase by Gαs. To determine the function of Gαs/microtubule association in neuronal development, we used real-time trafficking of a GFP-Gαs fusion protein. GFP-Gαs concentrates at the distal end of the neurites in differentiated living PC12 cells as well as in cultured hippocampal neurons. Gαs translocates to specialized membrane compartments at tips of growing neurites. A dominant-negative Gα chimera that interferes with Gαs binding to tubulin and activation of tubulin GTPase attenuates neurite elongation and neurite number both in PC12 cells and primary hippocampal neurons. This effect is greatest on differentiation induced by activated Gαs. Together, these data suggest that activated Gαs translocates from the plasma membrane and, through interaction with tubulin/microtubules in the cytosol, is important for neurite formation, development, and outgrowth. Characterization of neuronal G protein dynamics and their contribution to microtubule dynamics is important for understanding the molecular mechanisms by which G protein-coupled receptor signaling orchestrates neuronal growth and differentiation.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Hipocampo/metabolismo , Microtúbulos/metabolismo , Fator de Crescimento Neural/metabolismo , Neuritos/metabolismo , Tubulina (Proteína)/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/crescimento & desenvolvimento , Microtúbulos/efeitos dos fármacos , Microtúbulos/ultraestrutura , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Neuritos/ultraestrutura , Neurogênese/genética , Células PC12 , Ligação Proteica , Transporte Proteico , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Tubulina (Proteína)/genética
3.
Mol Microbiol ; 78(5): 1232-45, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21091507

RESUMO

Enteric bacterial pathogens have evolved sophisticated strategies to evade host immune defences. Some pathogens deliver anti-inflammatory effector molecules into the host cell cytoplasm via a type III secretion system (T3SS). Enteropathogenic Escherichia coli (EPEC) inhibits inflammation by an undefined, T3SS-dependent mechanism. Two proteins encoded outside of the EPEC locus of enterocyte effacement (LEE) pathogenicity island, non-LEE-encoded effector H1 (NleH1) and H2 (NleH2), display sequence similarity to Shigella flexneri OspG, which inhibits activation of the pro-inflammatory transcription factor NF-κB. We hypothesized that the anti-inflammatory effects of EPEC were mediated by NleH1 and NleH2. In this study, we examined the effect of NleH1/H2 on the NF-κB pathway. We show that NleH1/H2 are secreted via the T3SS and that transfection of cells with plasmids harbouring nleH1 or nleH2 decreased IKK-ß-induced NF-κB activity and attenuated TNF-α-induced degradation of phospho-IκBα by preventing ubiquitination. Serum KC levels were higher in mice infected with ΔnleH1H2 than those infected with WT EPEC, indicating that NleH1/H2 dampen pro-inflammatory cytokine expression. ΔnleH1H2 was cleared more rapidly than WT EPEC while complementation of ΔnleH1H2 with either NleH1 or NleH2 prolonged colonization. Together, these data show that NleH1 and NleH2 function to dampen host inflammation and facilitate EPEC colonization during pathogenesis.


Assuntos
Escherichia coli Enteropatogênica/imunologia , Infecções por Escherichia coli/imunologia , Proteínas de Escherichia coli/imunologia , NF-kappa B/imunologia , Animais , Linhagem Celular , Escherichia coli Enteropatogênica/genética , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Células HEK293 , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibidor de NF-kappaB alfa , NF-kappa B/genética
4.
Lab Invest ; 90(8): 1152-68, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20479715

RESUMO

Shiga toxin (Stx) is implicated in the development of hemorrhagic colitis and hemolytic-uremic syndrome, but early symptoms of enterohemorrhagic Escherichia coli (EHEC) infection such as nonbloody diarrhea may be Stx independent. In this study, we defined the effects of EHEC, in the absence of Stx, on the intestinal epithelium using a murine model. EHEC colonization of intestines from two groups of antibiotic-free and streptomycin-treated C57Bl/6J mice were characterized and compared. EHEC colonized the cecum and colon more efficiently than the ileum in both groups; however, greater amounts of tissue-associated EHEC were detected in streptomycin-pretreated mice. Imaging of intestinal tissues of mice infected with bioluminescent EHEC further confirmed tight association of the bacteria with the cecum and colon. Greater numbers of EHEC were also cultured from stool samples obtained from streptomycin-pretreated mice, as compared with those that received no antibiotics. Transmission electron microscopy shows that EHEC infection leads to microvillous effacement of mouse colonocytes. Hematoxylin and eosin staining of the colonic tissues of infected mice revealed a slight increase in the number of lamina propria polymorphonuclear leukocytes. Transmucosal electrical resistance, a measure of epithelial barrier function, was reduced in the colonic tissues of infected animals. Increased mucosal permeability to 4- kDa FITC-dextran was also observed in the colonic tissues of infected mice. Immunofluorescence microscopy showed that EHEC infection resulted in redistribution of the tight junction (TJ) proteins occludin and claudin-3 and increased the expression of claudin-2, whereas ZO-1 localization remained unaltered. Quantitative real-time PCR showed that EHEC altered mRNA transcription of OCLN, CLDN2, and CLDN3. Most notably, claudin-2 expression was significantly increased and correlated with increased intestinal permeability. Our data indicate that C57Bl/6J mice serve as an in vivo model to study the physiological effects of EHEC infection on the intestinal epithelium and suggest that altered transcription of TJ proteins has a role in the increase in intestinal permeability.


Assuntos
Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Escherichia coli/metabolismo , Mucosa Intestinal/microbiologia , Proteínas de Membrana/metabolismo , Animais , Claudina-3 , Colo/metabolismo , Colo/microbiologia , Dextranos , Diarreia , Escherichia coli Êntero-Hemorrágica , Escherichia coli/genética , Fluoresceína-5-Isotiocianato/análogos & derivados , Síndrome Hemolítico-Urêmica , Íleo/metabolismo , Mucosa Intestinal/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Ocludina , Permeabilidade , Toxina Shiga/metabolismo , Junções Íntimas/metabolismo , Junções Íntimas/microbiologia , Junções Íntimas/fisiologia
5.
Gut Microbes ; 1(2): 114-118, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21326920

RESUMO

Enteropathogenic Escherichia coli (EPEC) is an enteric pathogen able to cause severe diarrhea. Once adhered to the small intestine, EPEC disrupts tight junctions that are important for intestinal barrier function. This disruption is dependent on the bacterial type III secretion system, as well as the translocated effectors EspF and Map. Recently we have shown that a third type III translocated bacterial effector protein, NleA, is also involved in tight junction disruption during EPEC infection. NleA has a predicted PDZ-binding domain at its C-terminus which is proposed to be involved in protein interactions with PDZ domain containing proteins. Since several PDZ-domain-containing proteins localize to tight junctions, we hypothesized that the PDZ-binding domain of NleA might be important for its role in tight junction disruption. However, here we show that a molecular variant of NleA lacking the PDZ-binding domain behaves indistinguishably from the wild-type protein with respect to disruption of tight junctions.

6.
Cell Microbiol ; 12(1): 31-41, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19712078

RESUMO

Enteropathogenic Escherichia coli (EPEC) is a diarrhoeal pathogen that adheres to epithelial cells of the small intestine and uses a type III secretion system to inject effector proteins into host cells. EPEC infection leads to disruption of host intestinal tight junctions that are important for maintaining intestinal barrier function. This disruption is dependent on the bacterial type III secretion system, as well as the translocated effectors EspF and Map. Here we show that a third type III translocated bacterial effector protein, NleA, is also involved in tight junction disruption during EPEC infection. Using the drug Brefeldin A, we demonstrate that the effect of NleA on tight junction integrity is related to its inhibition of host cell protein trafficking through COPII-dependent pathways. These results suggest that NleA's striking effect on virulence is mediated, at least in part, via its role in disruption of intestinal barrier function.


Assuntos
Escherichia coli Enteropatogênica/fisiologia , Escherichia coli Enteropatogênica/patogenicidade , Proteínas de Escherichia coli/fisiologia , Intestinos/microbiologia , Junções Íntimas/metabolismo , Fatores de Virulência/fisiologia , Virulência/fisiologia , Brefeldina A/farmacologia , Células CACO-2 , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Linhagem Celular Tumoral , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Teste de Complementação Genética , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Microscopia de Fluorescência , Técnicas do Sistema de Duplo-Híbrido , Virulência/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
7.
Am J Physiol Gastrointest Liver Physiol ; 297(3): G576-81, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19556613

RESUMO

Infection with the enteric pathogen enterohemorrhagic Escherichia coli (EHEC) causes a variety of symptoms ranging from nonbloody diarrhea to more severe sequelae including hemorrhagic colitis, altered sensorium and seizures, and even life-threatening complications, such as hemolytic uremic syndrome and thrombotic thrombocytopenic purpura. The more severe consequences of EHEC infection are attributable to the production of Shiga toxin (Stx) and its subsequent effects on the vasculature, which expresses high levels of the Stx receptor, Gb3. Interestingly, the intestinal epithelium does not express Gb3. Despite the lack of Gb3 receptor expression, intestinal epithelial cells translocate Stx. The effect of Stx on intestinal epithelial cells is controversial with some studies demonstrating induction of inflammation and others not. This may be difficult to resolve because EHEC expresses both proinflammatory molecules, such as flagellin, and factor(s) that dampen the inflammatory response of epithelial cells. The goal of our study was to define the effect of Stx on the inflammatory response of intestinal epithelial cells and to determine whether infection by EHEC modulates this response. Here we show that Stx is a potent inducer of the inflammatory response in intestinal epithelial cells and confirm that EHEC attenuates the induction of IL-8 by host-derived proinflammatory cytokines. More importantly, however, we show that infection with EHEC attenuates the inflammatory response by intestinal epithelial cells to its own toxin. We speculate that the ability of EHEC to dampen epithelial cell inflammatory responses to Stx and cytokines facilitates intestinal colonization.


Assuntos
Citocinas/metabolismo , Enterite/microbiologia , Escherichia coli Êntero-Hemorrágica/patogenicidade , Células Epiteliais/microbiologia , Infecções por Escherichia coli/microbiologia , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/microbiologia , Toxinas Shiga/metabolismo , Enterite/imunologia , Enterite/prevenção & controle , Escherichia coli Êntero-Hemorrágica/metabolismo , Células Epiteliais/imunologia , Infecções por Escherichia coli/imunologia , Células HT29 , Interações Hospedeiro-Patógeno , Humanos , Proteínas I-kappa B/metabolismo , Interleucina-1beta/metabolismo , Interleucina-8/metabolismo , Mucosa Intestinal/imunologia , Inibidor de NF-kappaB alfa , Transporte Proteico , Triexosilceramidas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
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
9.
Am J Physiol Gastrointest Liver Physiol ; 294(5): G1165-70, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18356531

RESUMO

Enteropathogenic Escherichia coli (EPEC) is a diarrheagenic pathogen that perturbs intestinal epithelial function. Many of the alterations in the host cells are mediated by effector molecules that are secreted directly into epithelial cells by the EPEC type III secretion system. The secreted effector molecule EspF plays a key role in redistributing tight junction proteins and altering epithelial barrier function. EspF has also been shown to localize to mitochondria and trigger membrane depolarization and eventual host cell death. The relationship, if any, between EspF-induced host cell death and epithelial barrier disruption is presently not known. Site-directed mutation of leucine 16 (L16E) of EspF impairs both mitochondrial localization and consequent host cell death. Although the mutation lies within a region critical for type III secretion, EspF(L16E) is secreted efficiently from EPEC. Despite its inability to promote cell death, EspF(L16E) was not impaired for tight junction alteration or barrier disruption. Consistent with this, the pan-caspase inhibitor Q-VD-OPH, despite reducing EPEC-induced host cell death, had no effect on infection-mediated barrier function alteration. Thus EPEC alters the epithelial barrier independent of its ability to induce host cell death.


Assuntos
Apoptose/fisiologia , Escherichia coli Enteropatogênica/fisiologia , Mucosa Intestinal/fisiologia , Junções Íntimas/fisiologia , Clorometilcetonas de Aminoácidos/farmacologia , Substituição de Aminoácidos , Apoptose/efeitos dos fármacos , Células CACO-2 , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Inibidores de Caspase , Linhagem Celular Tumoral , Inibidores de Cisteína Proteinase/farmacologia , Impedância Elétrica , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Proteínas de Escherichia coli/biossíntese , Proteínas de Escherichia coli/genética , Humanos , Mucosa Intestinal/microbiologia , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/metabolismo , Ocludina , Permeabilidade/efeitos dos fármacos , Transporte Proteico/fisiologia , Quinolinas/farmacologia , Proteínas Recombinantes/biossíntese , Junções Íntimas/microbiologia
10.
Infect Immun ; 75(5): 2316-24, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17339360

RESUMO

The diarrheagenic pathogen enteropathogenic Escherichia coli (EPEC) is responsible for significant infant mortality and morbidity, particularly in developing countries. EPEC pathogenesis relies on a type III secretion system-mediated transfer of virulence effectors into host cells. EPEC modulates host cell survival and inflammation, although the proximal signaling pathways have not been well defined. We therefore examined the effect of EPEC on the epidermal growth factor receptor (EGFR), a known upstream activator of both the prosurvival phosphoinositide 3-kinase/Akt and proinflammatory mitogen-activated protein (MAP) kinase pathways. EPEC induced the autophosphorylation of EGFR in intestinal epithelial cells within 15 min postinfection, with maximal phosphorylation being observed at 4 h. Filter-sterilized supernatants of EPEC cultures also stimulated EGFR phosphorylation, suggesting that a secreted component(s) contributes to this activity. EPEC-induced EGFR phosphorylation was blocked by the pharmacological inhibitor tyrphostin AG1478, as well as by EGFR-neutralizing antibodies. Inhibition of EGFR phosphorylation by AG1478 had no effect on bacterial adherence, actin recruitment to sites of attachment, or EPEC-induced epithelial barrier function alteration. EPEC-mediated Akt phosphorylation, however, was inhibited by both AG1478 and EGFR-neutralizing antibodies. Correspondingly, inhibition of EGFR activation increased the apoptosis/necrosis of infected epithelial cells. Inhibition of EGFR phosphorylation also curtailed EPEC-induced ERK1/2 (MAP kinase) phosphorylation and, correspondingly, the production of the proinflammatory cytokine interleukin-8 by infected epithelial cells. Our studies suggest that EGFR is a key proximal signaling molecule during EPEC pathogenesis.


Assuntos
Células Epiteliais/microbiologia , Receptores ErbB/metabolismo , Escherichia coli/patogenicidade , Intestinos/microbiologia , Apoptose , Células CACO-2 , Células Epiteliais/química , Células Epiteliais/metabolismo , Receptores ErbB/genética , Células HeLa , Humanos , Interleucina-8/metabolismo , Intestinos/química , Intestinos/citologia , Fosforilação , Transdução de Sinais , Tirosina
11.
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
12.
Gastroenterology ; 127(3): 859-69, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15362041

RESUMO

BACKGROUND & AIMS: Enteropathogenic Escherichia coli and enterohemorrhagic E. coli harbor highly homologous pathogenicity islands yet show key differences in their mechanisms of action. Both disrupt host intestinal epithelial tight junctions, but the effects of enteropathogenic E. coli are more profound than those of enterohemorrhagic E. coli. The basis for this is not understood. The atypical protein kinase C isoform, protein kinase C-zeta, associates with and regulates the tight junction complex. The aim of this study was to compare the role of protein kinase C-zeta in the disruption of tight junctions after infection with enteropathogenic E. coli and enterohemorrhagic E. coli. METHODS: Model intestinal epithelial monolayers infected by enteropathogenic E. coli or enterohemorrhagic E. coli were used for these studies. RESULTS: Neither bisindolylmaleimide nor Gö6976, which block several protein kinase C isoforms but not protein kinase C-zeta, protected against the decrease in transepithelial electrical resistance after enteropathogenic E. coli infection. Rottlerin at concentrations that block novel and atypical isoforms, including protein kinase C-zeta, significantly attenuated the decrease in transepithelial electrical resistance. The specific inhibitory peptide, myristoylated protein kinase C-zeta pseudosubstrate, also significantly decreased the enteropathogenic E. coli -associated decrease in transepithelial electrical resistance and redistribution of tight junction proteins. In contrast to enteropathogenic E. coli, the level of protein kinase C-zeta enzyme activity stimulated by enterohemorrhagic E. coli was transient and minor, and protein kinase C-zeta inhibition had no effect on the decrease in transepithelial electrical resistance or the redistribution of occludin. CONCLUSIONS: The differential regulation of protein kinase C-zeta by enteropathogenic E. coli and enterohemorrhagic E. coli may in part explain the less profound effect of the latter on the barrier function of tight junctions.


Assuntos
Permeabilidade da Membrana Celular/fisiologia , Infecções por Escherichia coli/fisiopatologia , Escherichia coli/patogenicidade , Mucosa Intestinal/fisiopatologia , Proteína Quinase C/fisiologia , Junções Íntimas/fisiologia , Células Cultivadas , Células Epiteliais/fisiologia , Escherichia coli O157/patogenicidade , Humanos , Isoformas de Proteínas
13.
Cell Microbiol ; 6(10): 987-97, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15339273

RESUMO

Enteropathogenic Escherichia coli (EPEC) pathogenesis requires the delivery of effector proteins into host cytosol by a type III secretion system. The effector protein EspF, while critical for disruption of epithelial barrier function through alteration of tight junctions, is not required for bacterial viability or attachment. Yeast two-hybrid analyses revealed host intermediate filament (IF) protein cytokeratin 18 (CK18) as an interacting partner of EspF. This was confirmed by co-immunoprecipitation of extracts from EPEC-infected epithelial cells. EPEC infection increased detergent-soluble CK18 amounts without significantly altering CK18 expression. The adaptor protein 14-3-3 binds to CK18 and modulates its solubility. EPEC infection promoted CK18/14-3-3 interactions, corresponding to the increase of CK18 in the soluble fractions. 14-3-3 also co-immunoprecipitated with EspF, suggesting that CK18, 14-3-3 and EspF may form a complex in infected cells. The 14-3-3zeta isoform was co-immunoprecipitated with CK18, suggesting the involvement of specific signalling pathways. Immunofluorescence studies revealed a dramatic alteration in the architecture of the IF network in EPEC-infected epithelial cells. IF fragmentation, evident at 2 h post infection, progressed to a collapse of this network at later time points. The secretion mutant (DeltaescN) failed to alter CK18 solubility and IF morphology, while deletion of espF partially impaired the ability of EPEC to induce CK18 modifications. These results suggest that modifications in 14-3-3 interactions and IF network, modulated by type III secreted proteins, may be an important step in EPEC pathogenesis.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Queratinas/metabolismo , Proteínas 14-3-3/metabolismo , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Proteínas de Escherichia coli/genética , Humanos , Filamentos Intermediários/metabolismo , Técnicas do Sistema de Duplo-Híbrido
14.
Infect Immun ; 72(6): 3218-27, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15155623

RESUMO

Enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) are related intestinal pathogens that harbor highly similar pathogenicity islands known as the locus of enterocyte effacement (LEE). Despite their genetic similarity, these two pathogens disrupt epithelial tight junction barrier function with distinct kinetics. EHEC-induced reduction in transepithelial electrical resistance (TER), a measure of barrier function disruption, is significantly slower and more modest in comparison to that induced by EPEC. The variation in bacterial adherence only partially accounted for these differences. The LEE-encoded effector protein EspF has been shown to be critical for EPEC-induced alterations in TER. EspF from both EPEC and EHEC is expressed and secreted upon growth in tissue culture medium. The mutation of EHEC cesF suggested that the optimal expression and secretion of EHEC EspF required its chaperone CesF, as has been shown for EPEC. In contrast to EPEC espF and cesF, mutation of the corresponding EHEC homologs did not dramatically alter the decrease in TER. These differences could possibly be explained by the presence of additional espF-like sequences (designated U- and M-espF, where the letter designations refer to the specific cryptic prophage sequences on the EHEC chromosome closest to the respective genes) in EHEC. Reverse transcription-PCR analyses revealed coordinate regulation of EHEC U-espF and the LEE-encoded espF, with enhanced expression in bacteria grown in Dulbecco-Vogt modified Eagle's medium compared to bacteria grown in Luria broth. Both EHEC espF and U-espF complemented an EPEC espF deletion strain for barrier function alteration. The overexpression of U-espF, but not espF, in wild-type EHEC potentiated the TER response. These studies reveal further similarities and differences in the pathogenesis of EPEC and EHEC.


Assuntos
Células Epiteliais/patologia , Proteínas de Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Intestinos/patologia , Junções Íntimas/patologia , Aderência Bacteriana , Células CACO-2 , Linhagem Celular Tumoral , Células Epiteliais/citologia , Escherichia coli/classificação , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Humanos , Intestinos/citologia , Intestinos/microbiologia , Microscopia de Fluorescência , Chaperonas Moleculares/metabolismo , Junções Íntimas/fisiologia , Virulência
15.
Infect Immun ; 71(12): 7069-78, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14638797

RESUMO

Enteropathogenic Escherichia coli (EPEC) disrupts the structure and barrier function of host intestinal epithelial tight junctions (TJs). The impact of EPEC on TJ "fence function," i.e., maintenance of cell polarity, has not been investigated. In polarized cells, proteins such as beta(1)-integrin and Na(+)/K(+) ATPase are restricted to basolateral (BL) membranes. The outer membrane EPEC protein intimin possesses binding sites for the EPEC translocated intimin receptor (Tir) and beta(1)-integrin. Restriction of beta(1)-integrin to BL domains, however, precludes opportunity for interaction. We hypothesize that EPEC perturbs TJ fence function and frees BL proteins such as beta(1)-integrin to migrate to apical (AP) membranes of host cells, thus allowing interactions with bacterial adhesins such as intimin. The aim of this study was to determine whether EPEC alters the polar distribution of BL proteins, in particular beta(1)-integrin, and if such redistribution contributes to pathogenesis. Human intestinal epithelial T84 cells and EPEC strain E2348/69 were used. Selective biotinylation of AP or BL membrane proteins and confocal microscopy showed the presence of beta(1)-integrin and Na(+)/K(+) ATPase on the AP membrane following infection. beta(1)-Integrin antibody afforded no protection against the initial EPEC-induced decrease in transepithelial electrical resistance (TER) but halted the progressive decrease at later time points. While the effects of EPEC on TJ barrier and fence function were Tir dependent, disruption of cell polarity by calcium chelation allowed a tir mutant to be nearly as effective as wild-type EPEC. In contrast, deletion of espD, which renders the type III secretory system ineffective, had no effect on TER even after calcium chelation, suggesting that the putative beta(1)-integrin-intimin interaction serves to provide intimate contact, like that of Tir and intimin, making translocation of effector molecules more efficient. We conclude that the initial alterations of TJ barrier and fence function by EPEC are Tir dependent but that later disruption of cell polarity and accessibility of EPEC to BL membrane proteins, such as beta(1)-integrin, potentiates the physiological perturbations.


Assuntos
Membrana Celular/patologia , Polaridade Celular , Escherichia coli/patogenicidade , Junções Íntimas/patologia , Membrana Celular/fisiologia , Polaridade Celular/fisiologia , Células Cultivadas , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Proteínas de Escherichia coli , Humanos , Integrina beta1/metabolismo , Intestinos/citologia , Proteínas de Membrana/metabolismo , Microscopia Confocal , Receptores de Superfície Celular , ATPase Trocadora de Sódio-Potássio/metabolismo
16.
Am J Physiol Cell Physiol ; 285(3): C512-21, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12900386

RESUMO

We showed previously that enteropathogenic Escherichia coli (EPEC) infection of intestinal epithelial cells induces inflammation by activating NF-kappa B and upregulating IL-8 expression. We also reported that extracellular signal-regulated kinases (ERKs) participate in EPEC-induced NF-kappa B activation but that other signaling molecules such as PKC zeta may be involved. The aim of this study was to determine whether PKC zeta is activated by EPEC and to investigate whether it also plays a role in EPEC-associated inflammation. EPEC infection induced the translocation of PKC zeta from the cytosol to the membrane and its activation as determined by kinase activity assays. Inhibition of PKC zeta by the pharmacological inhibitor rottlerin, the inhibitory myristoylated PKC zeta pseudosubstrate (MYR-PKC zeta-PS), or transient expression of a nonfunctional PKC zeta significantly suppressed EPEC-induced I kappa B alpha phosphorylation. Although PKC zeta can activate ERK, MYR-PKC zeta-PS had no effect on EPEC-induced stimulation of this pathway, suggesting that they are independent events. PKC zeta can regulate NF-kappa B activation by interacting with and activating I kappa B kinase (IKK). Coimmunoprecipitation studies showed that the association of PKC zeta and IKK increased threefold 60 min after infection. Kinase activity assays using immunoprecipitated PKC zeta-IKK complexes from infected intestinal epithelial cells and recombinant I kappa B alpha as a substrate showed a 2.5-fold increase in I kappa B alpha phosphorylation. PKC zeta can also regulate NF-kappa B by serine phosphorylation of the p65 subunit. Serine phosphorylation of p65 was increased after EPEC infection but could not be consistently attenuated by MYR-PKC zeta-PS, suggesting that other signaling events may be involved in this particular arm of NF-kappa B regulation. We speculate that EPEC infection of intestinal epithelial cells activates several signaling pathways including PKC zeta and ERK that lead to NF-kappa B activation, thus ensuring the proinflammatory response.


Assuntos
Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/metabolismo , Proteína Quinase C/metabolismo , Células CACO-2 , Enterite/imunologia , Enterite/metabolismo , Células Epiteliais/enzimologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Humanos , Proteínas I-kappa B/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Transdução de Sinais/imunologia
17.
Gastroenterology ; 123(1): 163-72, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12105845

RESUMO

BACKGROUND & AIMS: Maintenance of the mucosal barrier is a critical function of intestinal epithelia. Myosin regulatory light chain (MLC) phosphorylation is a common intermediate in the pathophysiologic regulation of this barrier. The aim of this study was to determine whether a membrane permeant inhibitor of MLC kinase (PIK) could inhibit intracellular MLC kinase and regulate paracellular permeability. METHODS: Recombinant MLC and Caco-2 MLC kinase were used for kinase assays. T84 and Caco-2 monolayers were treated with enteropathogenic Escherichia coli (EPEC) or tumor necrosis factor (TNF)-alpha and interferon (IFN)-gamma to induce barrier dysfunction. RESULTS: PIK inhibited MLC kinase in vitro and was able to cross cell membranes and concentrate at the perijunctional actomyosin ring. Consistent with these properties, apical addition of PIK reduced intracellular MLC phosphorylation by 22% +/- 2%, increased transepithelial resistance (TER) by 50% +/- 1%, and decreased paracellular mannitol flux rates by 5.2 +/- 0.2-fold. EPEC infection induced TER decreases of 37% +/- 6% that were limited to 16% +/- 5% by PIK. TNF-alpha and IFN-gamma induced TER decreases of 22% +/- 3% that were associated with a 172% +/- 1% increase in MLC phosphorylation. Subsequent PIK addition caused MLC phosphorylation to decrease by 25% +/- 4% while TER increased to 97% +/- 6% of control. CONCLUSIONS: PIK can prevent TER defects induced by EPEC and reverse MLC phosphorylation increases and TER decreases induced by TNF-alpha and IFN-gamma. The data also suggest that TNF-alpha and IFN-gamma regulate TER, at least in part, via the perijunctional cytoskeleton. Thus, PIK may be the prototype for a new class of targeted therapeutic agents that can restore barrier function in intestinal disease states.


Assuntos
Inibidores Enzimáticos/farmacologia , Enteropatias/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Oligopeptídeos/farmacologia , Actomiosina/metabolismo , Sequência de Aminoácidos , Células CACO-2 , Membrana Celular/metabolismo , Inibidores Enzimáticos/farmacocinética , Infecções por Escherichia coli/metabolismo , Humanos , Interferon gama/farmacologia , Enteropatias/microbiologia , Enteropatias/patologia , Mucosa Intestinal/patologia , Membranas Intracelulares/metabolismo , Oligopeptídeos/farmacocinética , Permeabilidade , Fosforilação/efeitos dos fármacos , Junções Íntimas/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
18.
Infect Immun ; 70(5): 2271-7, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11953359

RESUMO

Disruption of the barrier properties of the enterocyte tight junction is believed to be important in the pathogenesis of diarrhea caused by enteropathogenic Escherichia coli (EPEC). This phenotype can be measured in vitro as the ability of EPEC to reduce transepithelial resistance (TER) across enterocyte monolayers and requires the products of the locus of enterocyte effacement (LEE) and, in particular, the type III secreted effector protein EspF. We report a second LEE-encoded gene that is also necessary for EPEC to fully reduce TER. rorf10 is not necessary for EPEC adherence, EspADB secretion, or formation of attaching and effacing lesions. However, rorf10 mutants have a diminished TER phenotype, reduced intracellular levels of EspF, and a reduced ability to translocate EspF into epithelial cells. The product of rorf10 is a 14-kDa intracellular protein rich in alpha-helices that specifically interacts with EspF but not with Tir or other EPEC secreted proteins. These properties are consistent with the hypothesis that rorf10 encodes a type III secretion chaperone for EspF, and we rename this protein CesF, the chaperone for EPEC secreted protein F.


Assuntos
Proteínas de Bactérias/metabolismo , Enterócitos/metabolismo , Escherichia coli/genética , Chaperonas Moleculares/genética , Junções Íntimas/metabolismo , Sequência de Aminoácidos , Transporte Biológico , Mapeamento Cromossômico , Escherichia coli/metabolismo , Células HeLa , Humanos , Dados de Sequência Molecular , Permeabilidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...