Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Pathol ; 174(4): 1347-57, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19286565

RESUMEN

Lymphocyte inhibitory factor A (lifA) in Citrobacter rodentium encodes the large toxin lymphostatin, which contains two enzymatic motifs associated with bacterial pathogenesis, a glucosyltransferase and a protease. Our aim was to determine the effects of each lymphostatin motif on intestinal epithelial-barrier function. In-frame mutations of C. rodentium lifA glucosyltransferase (CrGlM21) and protease (CrPrM5) were generated by homologous recombination. Infection of both model intestinal epithelial monolayers and mice with C. rodentium wild type resulted in compromised epithelial barrier function and mislocalization of key intercellular junction proteins in the tight junction and adherens junction. In contrast, CrGlM21 was impaired in its ability to reduce barrier function and influenced the tight junction proteins ZO-1 and occludin. CrPrM5 demonstrated decreased effects on the adherens junction proteins beta-catenin and E-cadherin. Analysis of the mechanisms revealed that C. rodentium wild type differentially influenced Rho GTPase activation, suppressed Cdc42 activation, and induced Rho GTPase activation. CrGlM21 lost its suppressive effects on Cdc42 activation, whereas CrPrM5 was unable to activate Rho signaling. Rescue experiments using constitutively active Cdc42 or C3 exotoxin to inhibit Rho GTPase supported a role of Rho GTPases in the epithelial barrier compromise induced by C. rodentium. Taken together, our results suggest that lymphostatin is a bacterial virulence factor that contributes to the disruption of intestinal epithelial-barrier function via the modulation of Rho GTPase activities.


Asunto(s)
Mucosa Intestinal/inmunología , Factores de Virulencia/inmunología , Proteínas de Unión al GTP rho/inmunología , Uniones Adherentes/metabolismo , Uniones Adherentes/patología , Animales , Citrobacter rodentium , Infecciones por Enterobacteriaceae/enzimología , Infecciones por Enterobacteriaceae/inmunología , Activación Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Glucosiltransferasas/genética , Glucosiltransferasas/inmunología , Mucosa Intestinal/enzimología , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Mutación , Péptido Hidrolasas/genética , Péptido Hidrolasas/inmunología , Factores de Virulencia/genética , Proteínas de Unión al GTP rho/metabolismo
2.
Am J Pathol ; 174(2): 436-48, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19147824

RESUMEN

Epithelial cell motility is critical for self-rejuvenation of normal intestinal mucosa, wound repair, and cancer metastasis. This process is regulated by the reorganization of the F-actin cytoskeleton, which is driven by a myosin II motor. However, the role of myosin II in regulating epithelial cell migration remains poorly understood. This study addressed the role of non-muscle myosin (NM) IIA in two different modes of epithelial cell migration: two-dimensional (2-D) migration that occurs during wound closure and three-dimensional (3-D) migration through a Matrigel matrix that occurs during cancer metastasis. Pharmacological inhibition or siRNA-mediated knockdown of NM IIA in SK-CO15 human colonic epithelial cells resulted in decreased 2-D migration and increased 3-D invasion. The attenuated 2-D migration was associated with increased cell adhesiveness to collagen and laminin and enhanced expression of beta1-integrin and paxillin. On the 2-D surface, NM IIA-deficient SK-CO15 cells failed to assemble focal adhesions and F-actin stress fibers. In contrast, the enhanced invasion of NM IIA-depleted cells was dependent on Raf-ERK1/2 signaling pathway activation, enhanced calpain activity, and increased calpain-2 expression. Our findings suggest that NM IIA promotes 2-D epithelial cell migration but antagonizes 3-D invasion. These observations indicate multiple functions for NM IIA, which, along with the regulation of the F-actin cytoskeleton and cell-matrix adhesions, involve previously unrecognized control of intracellular signaling and protein expression.


Asunto(s)
Adhesión Celular/fisiología , Movimiento Celular/fisiología , Mucosa Intestinal/metabolismo , Miosina Tipo IIA no Muscular/metabolismo , Transducción de Señal/fisiología , Animales , Western Blotting , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Colágeno , Combinación de Medicamentos , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Laminina , Microscopía Confocal , Proteoglicanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cicatrización de Heridas/fisiología
3.
J Immunol ; 181(7): 5035-44, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18802107

RESUMEN

During mucosal inflammation, a complex array of proinflammatory and protective mechanisms regulates inflammation and severity of injury. Secretion of anti-inflammatory mediators is a mechanism that is critical in controlling inflammatory responses and promoting epithelial restitution and barrier recovery. AnxA1 is a potent anti-inflammatory protein that has been implicated to play a critical immune regulatory role in models of inflammation. Although AnxA1 has been shown to be secreted in intestinal mucosal tissues during inflammation, its potential role in modulating the injury/inflammatory response is not understood. In this study, we demonstrate that AnxA1-deficient animals exhibit increased susceptibility to dextran sulfate sodium (DSS)-induced colitis with greater clinical morbidity and histopathologic mucosal injury. Furthermore, impaired recovery following withdrawal of DSS administration was observed in AnxA1 (-/-) animals compared with wild-type (WT) control mice that was independent of inflammatory cell infiltration. Since AnxA1 exerts its anti-inflammatory properties through stimulation of ALX/FPRL-1, we explored the role of this receptor-ligand interaction in regulating DSS-induced colitis. Interestingly, treatment with an ALX/FPRL-1 agonist, 15-epi-lipoxin A4 reversed the enhanced sensitivity of AnxA1 (-/-) mice to DSS colitis. In contrast, 15-epi-lipoxin A4 did not significantly improve the severity of disease in WT animals. Additionally, differential expression of ALX/FPLR-1 in control and DSS-treated WT and AnxA1-deficient animals suggested a potential role for AnxA1 in regulating ALX/FPRL-1 expression under pathophysiological conditions. Together, these results support a role of endogenous AnxA1 in the protective and reparative properties of the intestinal mucosal epithelium.


Asunto(s)
Anexina A1/fisiología , Mediadores de Inflamación/fisiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Cicatrización de Heridas/inmunología , Animales , Anexina A1/biosíntesis , Anexina A1/deficiencia , Anexina A1/genética , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/patología , Sulfato de Dextran/toxicidad , Femenino , Predisposición Genética a la Enfermedad , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Índice de Severidad de la Enfermedad , Cicatrización de Heridas/efectos de los fármacos
4.
J Cell Sci ; 121(11): 1803-14, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18460584

RESUMEN

The development of luminal organs begins with the formation of spherical cysts composed of a single layer of epithelial cells. Using a model three-dimensional cell culture, this study examines the role of a cytoskeletal motor, myosin II, in cyst formation. Caco-2 and SK-CO15 intestinal epithelial cells were embedded into Matrigel, and myosin II was inhibited by blebbistatin or siRNA-mediated knockdown. Whereas control cells formed spherical cysts with a smooth surface, inhibition of myosin II induced the outgrowth of F-actin-rich surface protrusions. The development of these protrusions was abrogated after inhibition of F-actin polymerization or of phospholipase C (PLC) activity, as well as after overexpression of a dominant-negative ADF/cofilin. Surface protrusions were enriched in microtubules and their formation was prevented by microtubule depolymerization. Myosin II inhibition caused a loss of peripheral F-actin bundles and a submembranous extension of cortical microtubules. Our findings suggest that inhibition of myosin II eliminates the cortical F-actin barrier, allowing microtubules to reach and activate PLC at the plasma membrane. PLC-dependent stimulation of ADF/cofilin creates actin-filament barbed ends and promotes the outgrowth of F-actin-rich protrusions. We conclude that myosin II regulates the spherical shape of epithelial cysts by controlling actin polymerization at the cyst surface.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Forma de la Célula/fisiología , Células Epiteliales/metabolismo , Mucosa Intestinal/embriología , Mucosa Intestinal/metabolismo , Miosina Tipo II/metabolismo , Actinas/metabolismo , Células CACO-2 , Polaridad Celular/fisiología , Forma de la Célula/efectos de los fármacos , Cofilina 1/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Mucosa Intestinal/citología , Miosina Tipo II/genética , Nocodazol/farmacología , Técnicas de Cultivo de Órganos , Polímeros/metabolismo , Estabilidad del ARN , Fosfolipasas de Tipo C/metabolismo
5.
Gastroenterology ; 134(1): 120-30, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18054006

RESUMEN

BACKGROUND & AIMS: Krüppel-like factor 5 (KLF5) is a zinc finger-transcription factor that regulates cell proliferation. Oncogenic KRAS mutations are commonly found in colorectal cancers. We aimed to determine whether KLF5 mediates KRAS functions during intestinal tumorigenesis. METHODS: The effects of KLF5 on proliferation and transformation were examined in IEC-6 intestinal epithelial cells stably transfected with inducible KRAS(V12G). KLF5 expression was examined in intestinal tumors derived from transgenic mice expressing KRAS(V12G) under villin promoter and in human colorectal cancers with mutated KRAS. RESULTS: Induction of KRAS(V12G) in IEC-6 cells resulted in increased expression of KLF5, accompanied by increased rates of proliferation and anchorage-independent growth. Inhibition of KLF5 expression by mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) inhibitors or KLF5-specific small interfering RNA reduced proliferation and anchorage-independent growth despite KRAS(V12G) induction. Human colorectal cancer cell lines with mutated KRAS contained high levels of KLF5 and reduction of KLF5 by MEK inhibitors or KLF5 small interfering RNA also led to reduced proliferation and transformation. In vivo, both intestinal tumors derived from mice transgenic for villin-KRAS(V12G) and human primary colorectal cancers with mutated KRAS contained high levels of KLF5 and increased staining of the proliferative marker Ki67. CONCLUSIONS: Elevated levels of KLF5 protein are strongly correlated with activating KRAS mutations in intestinal tumors in vitro and in vivo. Inhibition of KLF5 expression in tumor cells resulted in significantly reduced rates of proliferation and transforming activities. We conclude that KLF5 is an important mediator of oncogenic KRAS transforming functions during intestinal tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/patología , Neoplasias Colorrectales/etiología , Células Epiteliales/fisiología , Mucosa Intestinal/patología , Factores de Transcripción de Tipo Kruppel/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas ras/fisiología , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/patología , Humanos , Ratones , Proteínas Proto-Oncogénicas p21(ras)
6.
J Immunol ; 179(12): 8112-21, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18056353

RESUMEN

Inflammatory disorders of the gastrointestinal tract result in the breakdown of the intestinal epithelial barrier in the form of erosion and ulceration. To reestablish the epithelial barrier, the epithelium must efficiently migrate to reseal wounds. Numerous signaling cascades are involved in the induction and regulation of this complex process. N-formyl peptide receptors comprise a group of Gi-coupled receptors that regulate innate immune responses. Previously, we identified the expression of functional N-formyl peptide receptors in model SK-CO15 intestinal epithelial cells and observed a role for activation of these receptors in regulating cellular invasive behavior. In these studies, we performed formyl peptide receptor-1 (FPR) localization and evaluated its role in regulating intestinal epithelial cell wound closure. Immunolocalization studies using a recently developed specific monoclonal anti-FPR Ab demonstrated its localization along the lateral membrane of crypt epithelial cells in normal human colonic epithelium. In vitro studies using the classical FPR agonist fMLF showed that FPR activation significantly enhances model intestinal epithelial cell restitution and that FPR localized along actin filaments in lamellipodial and filopodial extrusions. The increase in cell migration was associated with activation of PI3K, Rac1, and Cdc42. Pharmacologic inhibition of PI3K activity abrogated the fMLF-induced increase in wound closure and activation of both Rac1 and Cdc42. Inhibition of Rac1 and Cdc42 using pharmacologic inhibitors and dominant negative mutants also inhibited the fMLF-induced increase in cell migration. Taken together, theses results support a novel role for FPR stimulation in enhancing intestinal epithelial cell restitution through PI3K-dependent activation of Rac1 and Cdc42.


Asunto(s)
Mucosa Intestinal/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Formil Péptido/fisiología , Cicatrización de Heridas , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/lesiones , Inhibidores de las Quinasa Fosfoinosítidos-3 , Receptores de Formil Péptido/agonistas , Receptores de Formil Péptido/análisis
7.
J Exp Med ; 204(13): 3067-76, 2007 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-18039951

RESUMEN

Recent evidence has linked intestinal permeability to mucosal inflammation, but molecular studies are lacking. Candidate regulatory molecules localized within the tight junction (TJ) include Junctional Adhesion Molecule (JAM-A), which has been implicated in the regulation of barrier function and leukocyte migration. Thus, we analyzed the intestinal mucosa of JAM-A-deficient (JAM-A(-/-)) mice for evidence of enhanced permeability and inflammation. Colonic mucosa from JAM-A(-/-) mice had normal epithelial architecture but increased polymorphonuclear leukocyte infiltration and large lymphoid aggregates not seen in wild-type controls. Barrier function experiments revealed increased mucosal permeability, as indicated by enhanced dextran flux, and decreased transepithelial electrical resistance in JAM-A(-/-) mice. The in vivo observations were epithelial specific, because monolayers of JAM-A(-/-) epithelial cells also demonstrated increased permeability. Analyses of other TJ components revealed increased expression of claudin-10 and -15 in the colonic mucosa of JAM-A(-/-) mice and in JAM-A small interfering RNA-treated epithelial cells. Given the observed increase in colonic inflammation and permeability, we assessed the susceptibility of JAM-A(-/-) mice to the induction of colitis with dextran sulfate sodium (DSS). Although DSS-treated JAM-A(-/-) animals had increased clinical disease compared with controls, colonic mucosa showed less injury and increased epithelial proliferation. These findings demonstrate a complex role of JAM-A in intestinal homeostasis by regulating epithelial permeability, inflammation, and proliferation.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Colon/metabolismo , Mucosa Intestinal/metabolismo , Receptores de Superficie Celular/fisiología , Animales , Línea Celular Tumoral , Epitelio/embriología , Predisposición Genética a la Enfermedad , Humanos , Inflamación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Neutrófilos/metabolismo , Permeabilidad
8.
PLoS One ; 2(7): e658, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17668046

RESUMEN

The integrity and function of the epithelial barrier is dependent on the apical junctional complex (AJC) composed of tight and adherens junctions and regulated by the underlying actin filaments. A major F-actin motor, myosin II, was previously implicated in regulation of the AJC, however direct evidence of the involvement of myosin II in AJC dynamics are lacking and the molecular identity of the myosin II motor that regulates formation and disassembly of apical junctions in mammalian epithelia is unknown. We investigated the role of nonmuscle myosin II (NMMII) heavy chain isoforms, A, B, and C in regulation of epithelial AJC dynamics and function. Expression of the three NMMII isoforms was observed in model intestinal epithelial cell lines, where all isoforms accumulated within the perijunctional F-actin belt. siRNA-mediated downregulation of NMMIIA, but not NMMIIB or NMMIIC expression in SK-CO15 colonic epithelial cells resulted in profound changes of cell morphology and cell-cell adhesions. These changes included acquisition of a fibroblast-like cell shape, defective paracellular barrier, and substantial attenuation of the assembly and disassembly of both adherens and tight junctions. Impaired assembly of the AJC observed after NMMIIA knock-down involved dramatic disorganization of perijunctional actin filaments. These findings provide the first direct non-pharmacological evidence of myosin II-dependent regulation of AJC dynamics in mammalian epithelia and highlight a unique role of NMMIIA in junctional biogenesis.


Asunto(s)
Células Epiteliales/química , Uniones Intercelulares/química , Cadenas Pesadas de Miosina/análisis , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Línea Celular Tumoral/efectos de los fármacos , Polaridad Celular , Cartilla de ADN , Regulación hacia Abajo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Silenciador del Gen , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Uniones Intercelulares/efectos de los fármacos , Mucosa Intestinal/química , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Cadenas Pesadas de Miosina/genética , Reacción en Cadena de la Polimerasa , Isoformas de Proteínas/química , ARN Interferente Pequeño/genética
9.
Lab Invest ; 87(10): 1042-54, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17660846

RESUMEN

Crohn's disease (CD) and ulcerative colitis (UC) are idiopathic inflammatory conditions of the gut. Our goal was to investigate if invasive Escherichia coli strains were present in patients with inflammatory bowel disease (IBD). Bacterial strains were isolated from biopsy material obtained from normal controls, and patients with a clinical diagnosis of CD and UC. Invasive bacteria were characterized by gentamicin protection assay and biochemical profiling (Api-20E). Strains were characterized by induction of cytokine expression in epithelial and macrophage cell cultures, measurement of epithelial barrier function, and confocal microscopy. Of all invasive bacterial strains in CD 98.9% were identified as E. coli as opposed to 42.1% in UC and 2.1% in normal controls. Epithelial invasion in vitro was significantly higher for CD-associated E. coli (8.4%, +/-5.5 of initial inoculum (I/O)) in comparison to UC (2.5%, +/-0.4 I/O), but highest for strains from inflamed CD tissue (11.3%, +/-4.3 I/O). Both, CD and UC E. coli strains induced high mean TNF-alpha expression in macrophage cell lines (2604.8 pg/10(5) cells, +/-447.4; 2,402.6 pg/10(5) cells, +/-476.3, respectively), but concentrations were significantly higher for isolates from inflamed CD tissue (3071.3 pg/10(5) cells, +/-226.0). Invasive E. coli from IBD tissue induced similar concentrations of interleukin (IL)-8 in epithelial cell cultures, but strains from inflamed CD tissue induced significantly less epithelial IL-8 (674.1 pg/10(5) cells, +/-58.0 vs 920.5 pg/10(5) cells, +/-94.6). IBD-associated E. coli strains significantly decreased transepithelial resistance, induced disorganization of F-actin and displacement of ZO-1, and E-cadherin from the apical junctional complex (AJC). In comparison to normal controls and UC, E. coli are more prevalent in CD, are highly invasive, and do not encode for known effector proteins. E. coli strains from IBD patients regulate cytokine expression and epithelial barrier function, two pathological features of IBD.


Asunto(s)
Colitis Ulcerosa/microbiología , Enfermedad de Crohn/microbiología , Citocinas/metabolismo , Escherichia coli/patogenicidad , Mucosa Intestinal/microbiología , Células CACO-2 , Estudios de Casos y Controles , Colitis Ulcerosa/metabolismo , Enfermedad de Crohn/metabolismo , Escherichia coli/genética , Humanos , Mucosa Intestinal/metabolismo , Permeabilidad
10.
Am J Pathol ; 170(3): 951-66, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17322380

RESUMEN

Epithelial cell migration is a critical event in gastrointestinal mucosal wound healing and is dependent on actin cytoskeletal reorganization. We observed increased expression of an actin regulatory protein, annexin 2, in migrating intestinal epithelial cells. Small interfering RNA (siRNA)-mediated knockdown of annexin 2 expression in Caco-2 epithelial cells resulted in significant reductions in cell spreading and wound closure associated with decreased formation of filamentous actin bundles along the base of migrating cells. Because annexin 2 has been shown to influences actin cytoskeletal remodeling through targeting signaling molecules to membrane domains, we examined the membrane association and activation status of Rho GTPases after annexin 2 knockdown. We observed Rho dissociation from membranes and decreased Rho activity following annexin 2 siRNA transfection. Inhibition of cell spreading and wound closure in annexin 2 siRNA-transfected cells was prevented by expression of constitutively active RhoA. Rho colocalized with annexin 2 in lamellipodia and along the cytoplasmic face of the plasma membrane. In addition, annexin 2 was observed to co-immunoprecipitate with endogenous Rho and constitutively active RhoA. These findings suggest that annexin 2 plays a role in targeting Rho to cellular membranes, thereby modulating Rho-related signaling events regulating cytoskeletal reorganization during epithelial cell migration.


Asunto(s)
Anexina A2/metabolismo , Movimiento Celular/fisiología , Mucosa Intestinal/metabolismo , Transducción de Señal/fisiología , Cicatrización de Heridas/fisiología , Proteína de Unión al GTP rhoA/metabolismo , Actinas/metabolismo , Células CACO-2 , Células Epiteliales/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Procesamiento de Imagen Asistido por Computador , Immunoblotting , Inmunoprecipitación , Microscopía Confocal , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
11.
Am J Pathol ; 169(5): 1686-700, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17071592

RESUMEN

Flagellin, the primary component of bacterial flagella, is a potent activator of toll-like receptor 5 (TLR5) signaling and is a major proinflammatory determinant of enteropathogenic Salmonella. In accordance with this, we report here that aflagellate Salmonella mutants are impaired in their ability to up-regulate proinflammatory and anti-apoptotic effector molecules in murine models of salmonellosis and that these mutants elicit markedly reduced early mucosal inflammation relative to their isogenic parent strains. Conversely, aflagellate bacteria were more potent activators of epithelial caspases and subsequent apoptosis. These phenomena correlated with a delayed but markedly exacerbated mucosal inflammation at the later stages of infection as well as elevated extra-intestinal and systemic bacterial load, culminating in a more severe clinical outcome. Systemic administration of exogenous flagellin primarily reversed the deleterious effects of in vivo Salmonella infection. These observations indicate that in Salmonella infection, flagellin plays a dominant role in activation of not only innate immunity but also anti-apoptotic processes in epithelial cells. These latter TLR-mediated responses that delay epithelial apoptosis may be as critical to mucosal defense as the classic acute inflammatory response. This notion is consistent with the emerging paradigm that specific TLR ligands may have a fundamental cytoprotective effect during inflammatory stress.


Asunto(s)
Apoptosis , Células Epiteliales/patología , Flagelina/metabolismo , Infecciones por Salmonella/patología , Animales , Células CACO-2 , Caspasas/metabolismo , Ciego/citología , Ciego/microbiología , Ciego/patología , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Enteritis/patología , Femenino , Humanos , Íleon/citología , Íleon/microbiología , Íleon/patología , Inflamación , Mucosa Intestinal/citología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos BALB C , Ratas , Salmonella/patogenicidad
12.
Clin Gastroenterol Hepatol ; 4(10): 1193-8, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16979950

RESUMEN

A wide variety of small intestinal mucosal diseases lead to malabsorption. Although stool studies, especially stool for excess fat, and functional tests for deficiency states are important clues to malabsorption, small intestinal biopsies are probably the most crucial part of the diagnostic process. Many mucosal disorders have distinctive histologic features that allow for precise diagnosis. However, these histologic changes might be subtle. The role of the gastroenterologist is to provide the pathologist with adequate clinical information and tissue material to ensure a complete examination pathologically. Celiac disease is the most common mucosal cause of chronic malabsorption in the western world. Celiac disease can present classically as large volume fatty diarrhea, but it more commonly presents with subtle clinical symptoms or iron deficiency anemia. Although the histologic hallmark of celiac disease is increased intraepithelial lymphocytosis along with villous atrophy, increased intraepithelial lymphocytosis alone in an appropriate clinical context might suggest the diagnosis of celiac disease. The aim of this review is to highlight the importance of close cooperation and communication between the gastroenterologist and the pathologist to optimize the diagnosis of mucosal diseases that result in malabsorption.


Asunto(s)
Enfermedad Celíaca/patología , Intestino Delgado/patología , Adulto , Biopsia , Diagnóstico Diferencial , Femenino , Humanos
13.
J Biol Chem ; 281(28): 19588-99, 2006 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-16675446

RESUMEN

Annexin 1 (AnxA1) is a multifunctional phospholipid-binding protein associated with the development of metastasis in some invasive epithelial malignancies. However, the role of AnxA1 in the migration/invasion of epithelial cells is not known. In this study, experiments were performed to investigate the role of AnxA1 in the invasion of a model epithelial cell line, SKCO-15, derived from colorectal adenocarcinoma. Small interfering RNA-mediated knockdown of AnxA1 expression resulted in a significant reduction in invasion through Matrigel-coated filters. Localization studies revealed a translocation of AnxA1 to the cell surface upon the induction of cell migration, and functional inhibition of cell surface AnxA1 using antiserum (LCO1) significantly reduced cell invasion. Conversely, SKCO-15 cell invasion was increased by approximately 2-fold in the presence of recombinant full-length AnxA1 and the AnxA1 N-terminal-derived peptide mimetic, Ac2-26. Because extracellular AnxA1 has been shown to regulate leukocyte migratory events through interactions with n-formyl peptide receptors (nFPRs), we examined the expression of FPR-1, FPRL-1, and FPRL-2 in SKCO-15 cells by reverse transcriptase-PCR and identified expression of all three receptors in this cell line. Treatment of SKCO-15 cells with AnxA1, Ac2-26, and the classical nFPR agonist, formylmethionylleucylphenylalanine, induced intracellular calcium release consistent with nFPR activation. Furthermore, the nFPR antagonist, Boc2, abrogated the AnxA1 and Ac2-26-induced intracellular calcium release and increase in SKCO-15 cell invasion. Together, these results support an autocrine/paracrine role for membrane AnxA1 in stimulating SKCO-15 cell migration through nFPR activation. The findings in this study suggest that activation of nFPRs stimulates epithelial cell motility important in the development of metastasis as well as wound healing.


Asunto(s)
Anexina A1/fisiología , Células Epiteliales/metabolismo , Receptores de Formil Péptido/química , Anexina A1/metabolismo , Anexina A1/farmacología , Línea Celular Tumoral , Membrana Celular/metabolismo , Movimiento Celular , Humanos , N-Formilmetionina Leucil-Fenilalanina/farmacología , Invasividad Neoplásica , Péptidos/farmacología , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/química , Transducción de Señal , Factores de Tiempo , Cicatrización de Heridas
14.
Int J Pharm ; 315(1-2): 122-33, 2006 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-16564652

RESUMEN

This study was conducted to evaluate the ability of electroporation to efficiently transfect differentiated intestinal epithelial monolayers with plasmid DNA and to determine whether electroporation can transfect these monolayers with short-interfering RNA (siRNA) to cause gene silencing. Confluent T84 monolayers were transfected with reporter plasmids expressing luciferase or green-fluorescent protein or with siRNA directed against the nuclear envelope proteins lamin A/C using electroporation. Optimized electroporation conditions resulted in luciferase and GFP expression. Both intracellular uptake of fluorescently labeled plasmid and expression of the reporter genes increased with increasing electroporation strength and DNA concentration. When monolayers were transfected by lipofection with the reporter plasmids, expression and DNA uptake were less than for electroporation. Electroporation was also found to transfect monolayers with siRNA, which resulted in up to 90% inhibition of targeted protein production. Silencing occurred within 24h of transfection and increased with increasing siRNA concentration. These results suggest that electroporation can provide a valuable research tool for transfection of intestinal epithelial monolayers and other differentiated cell systems, and may ultimately be useful for clinical gene therapy applications.


Asunto(s)
Electroporación/métodos , Mucosa Intestinal/citología , Plásmidos/genética , ARN Interferente Pequeño/genética , Transfección/métodos , Línea Celular , Electroporación/normas , Células Epiteliales/metabolismo , Silenciador del Gen , Genes Reporteros , Humanos , Proteínas/antagonistas & inhibidores , Proteínas/genética , Transfección/normas
15.
BMC Cell Biol ; 7: 12, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16509970

RESUMEN

BACKGROUND: Epithelial tight junction (TJ) and adherens junction (AJ) form the apical junctional complex (AJC) which regulates cell-cell adhesion, paracellular permeability and cell polarity. The AJC is anchored on cytoskeletal structures including actin microfilaments and microtubules. Such cytoskeletal interactions are thought to be important for the assembly and remodeling of apical junctions. In the present study, we investigated the role of microtubules in disassembly of the AJC in intestinal epithelial cells using a model of extracellular calcium depletion. RESULTS: Calcium depletion resulted in disruption and internalization of epithelial TJs and AJs along with reorganization of perijunctional F-actin into contractile rings. Microtubules reorganized into dense plaques positioned inside such F-actin rings. Depolymerization of microtubules with nocodazole prevented junctional disassembly and F-actin ring formation. Stabilization of microtubules with either docetaxel or pacitaxel blocked contraction of F-actin rings and attenuated internalization of junctional proteins into a subapical cytosolic compartment. Likewise, pharmacological inhibition of microtubule motors, kinesins, prevented contraction of F-actin rings and attenuated disassembly of apical junctions. Kinesin-1 was enriched at the AJC in cultured epithelial cells and it also accumulated at epithelial cell-cell contacts in normal human colonic mucosa. Furthermore, immunoprecipitation experiments demonstrated association of kinesin-1 with the E-cadherin-catenin complex. CONCLUSION: Our data suggest that microtubules play a role in disassembly of the AJC during calcium depletion by regulating formation of contractile F-actin rings and internalization of AJ/TJ proteins.


Asunto(s)
Citoesqueleto de Actina/ultraestructura , Uniones Adherentes/ultraestructura , Células Epiteliales/ultraestructura , Microtúbulos/fisiología , Uniones Estrechas/ultraestructura , Actinas/análisis , Animales , Calcio/fisiología , Línea Celular , Perros , Humanos , Cinesinas/antagonistas & inhibidores , Cinesinas/fisiología , Microtúbulos/ultraestructura
17.
Mol Biol Cell ; 16(6): 2694-703, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15800062

RESUMEN

Neutrophil (polymorphonuclear leukocytes [PMN]) transepithelial migration during inflammatory episodes involves a complex series of adhesive interactions and signaling events. Previous studies have shown that key adhesive interactions between leukocyte CD11b/CD18 and basally expressed fucosylated glycoproteins followed by binding to desmosomal-associated JAM-C are key elements of the transmigration response. Here we provide the first evidence that PMN-expressed junctional adhesion molecule-like protein (JAML) regulates transmigration via binding interactions with epithelial coxsackie and adenovirus receptor (CAR). Experiments with a JAML fusion protein revealed specific binding of JAML to epithelial CAR expressed at tight junctions in T84 cell monolayers and normal human colonic mucosa. Furthermore, JAML-CAR binding is mediated via the membrane distal immunoglobulin (Ig) loop of CAR and the membrane proximal Ig loop of JAML. PMN bound to immobilized CAR but not JAML in a divalent cation-independent manner. Lastly, in assays of PMN transepithelial migration, JAML/CAR fusion proteins and their antibodies significantly inhibited transmigration in a specific manner. Taken together, these results indicate that JAML and CAR are a novel pair of adhesion molecules that play an important role in modulating PMN migration cross epithelial tight junctions. These findings add a new element to a multistep model of PMN transepithelial migration and may provide new targets for anti-inflammatory therapies.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Quimiotaxis de Leucocito , Células Epiteliales/metabolismo , Neutrófilos/fisiología , Receptores Virales/metabolismo , Uniones Estrechas/metabolismo , Anticuerpos Monoclonales/metabolismo , Adhesión Celular , Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/genética , Línea Celular , Colon/citología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Colorantes Fluorescentes , Glutatión Transferasa/metabolismo , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Moléculas de Adhesión de Unión , Modelos Biológicos , Neutrófilos/química , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Solubilidad
18.
Infect Immun ; 73(3): 1441-51, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15731042

RESUMEN

Previously, we have identified a large gene (lifA, for lymphocyte inhibitory factor A) in enteropathogenic Escherichia coli (EPEC) encoding a protein termed lymphostatin that suppresses cytokine expression in vitro. This protein also functions as an adhesion factor for enterohemorrhagic E. coli (EHEC) and Shiga toxin-producing E. coli and is alternatively known as efa1 (EHEC factor for adherence 1). The lifA/efa1 gene is also present in Citrobacter rodentium, an enteric pathogen that causes a disease termed transmissible murine colonic hyperplasia (TMCH), which induces colitis and massive crypt cell proliferation, in mice. To determine if lifA/efa1 is required for C. rodentium-induced colonic pathology in vivo, three in-frame mutations were generated, disrupting the glycosyltransferase (GlM12) and protease (PrMC31) motifs and a domain in between that does not encode any known activity (EID3). In contrast to infection with wild-type C. rodentium, that with any of the lifA/efa1 mutant strains did not induce weight loss or TMCH. Enteric infection with motif mutants GlM12 and PrM31 resulted in significantly reduced colonization counts during the entire 20-day course of infection. In contrast, EID3 was indistinguishable from the wild type during the initial colonic colonization, but cleared rapidly after day 8 of the infection. The colonic epithelium of all infected mice displayed increased epithelial regeneration. However, significantly increased regeneration was observed by day 20 only in mice infected with the wild-type in comparison to those infected with lifA/efa1 mutant EID3. In summary, lifA/efa1 is a critical gene outside the locus for enterocyte effacement that regulates bacterial colonization, crypt cell proliferation, and epithelial cell regeneration.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Citrobacter rodentium/patogenicidad , Colon/microbiología , Colon/patología , Proteínas de Escherichia coli/metabolismo , Animales , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Colitis/microbiología , Colon/citología , Proteínas de Escherichia coli/genética , Femenino , Regulación Bacteriana de la Expresión Génica , Humanos , Hiperplasia/microbiología , Lactante , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Mutación , Análisis de Secuencia de ADN
19.
J Biol Chem ; 280(12): 11665-74, 2005 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-15677455

RESUMEN

Epithelial tight junctions form a selectively permeable barrier to ions and small molecules. Junctional adhesion molecule 1 (JAM1/JAM-A/F11R) is a tight junction-associated transmembrane protein that has been shown to participate in the regulation of epithelial barrier function. In a recent study, we presented evidence suggesting that JAM1 homodimer formation is critical for epithelial barrier function (Mandell, K. J., McCall, I. C., and Parkos, C. A. (2004) J. Biol. Chem. 279, 16254-16262). Here we have used small interfering RNA to investigate the effect of the loss of JAM1 expression on epithelial cell function. Consistent with our previous study, knockdown of JAM1 was observed to increase paracellular permeability in epithelial monolayers. Interestingly, knockdown of JAM1 also produced dramatic changes in cell morphology, and a similar effect was observed with expression of a JAM1 mutant lacking the putative homodimer interface. Further studies revealed that JAM1 knockdown decreased cell-matrix adhesion and spreading on matrix proteins that are ligands of beta1 integrins. These changes were characterized by a decrease in beta1 integrin protein levels and loss of beta1 integrin staining at the cell surface. Immunolabeling of cells for the small GTPase Rap1, a known activator of beta1 integrins, revealed colocalization of Rap1 with JAM1 at intercellular junctions, and knockdown of JAM1 resulted in decreased Rap1 activity. Lastly, knockdown of Rap1b resulted in diminished beta1 integrin expression and altered cell morphology analogous to that observed with knockdown of JAM1. Together, these results suggest that JAM1 regulates epithelial cell morphology and beta1 integrin expression by modulating activity of the small GTPase Rap1.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Integrina beta1/fisiología , Receptores de Superficie Celular/fisiología , Proteínas de Unión al GTP rap1/fisiología , Adhesión Celular , Moléculas de Adhesión Celular/genética , Línea Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Permeabilidad , ARN Interferente Pequeño/farmacología , Receptores de Superficie Celular/genética
20.
Am J Physiol Gastrointest Liver Physiol ; 287(4): G886-98, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15191880

RESUMEN

Superficial wounds in the gastrointestinal tract rapidly reseal by coordinated epithelial cell migration facilitated by cytokines such as hepatocyte growth factor (HGF)/scatter factor released in the wound vicinity. However, the mechanisms by which HGF promotes physiological and pathophysiologic epithelial migration are incompletely understood. Using in vitro models of polarized T84 and Caco-2 intestinal epithelia, we report that HGF promoted epithelial spreading and RhoA GTPase activation in a time-dependent manner. Inducible expression of enhanced green fluorescent protein-tagged dominant-negative RhoA significantly attenuated HGF-induced spreading. HGF expanded a zone of partially flattened cells behind the wound edge containing basal F-actin fibers aligned in the direction of spreading. Concomitantly, plaques positive for the focal adhesion protein paxillin were enhanced. HGF induced an increase in the translation of paxillin and, to a lesser extent, beta1-integrin. This was independent of cell-matrix adhesion through beta1-integrin. Subcellular fractionation revealed increased cosedimentation of paxillin with plasma membrane-containing fractions following HGF stimulation, without corresponding enhancements in paxillin coassociation with beta1 integrin or actin. Tyrosine phosphorylation of paxillin was reduced by HGF and was sensitive to the Src kinase inhibitor PP2. With these taken together, we propose that HGF upregulates a free cytosolic pool of paxillin that is unaffiliated with either the cytoskeleton or focal cell-matrix contacts. Thus early spreading responses to HGF may partly relate to increased paxillin availability for incorporation into, and turnover within, dynamic cytoskeletal/membrane complexes whose rapid and transient adhesion to the matrix drives migration.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Fosfoproteínas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Actinas/metabolismo , Células CACO-2 , Membrana Celular/metabolismo , Movimiento Celular/fisiología , Células Cultivadas , Colon/citología , Citosol/metabolismo , Células Epiteliales/citología , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/fisiología , Humanos , Integrina beta1/metabolismo , Paxillin , Fosforilación/efectos de los fármacos , Regulación hacia Arriba , Proteína de Unión al GTP rhoA/metabolismo , Familia-src Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA