Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Am J Pathol ; 174(4): 1347-57, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19286565

RESUMO

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.


Assuntos
Mucosa Intestinal/imunologia , Fatores de Virulência/imunologia , Proteínas rho de Ligação ao GTP/imunologia , Junções Aderentes/metabolismo , Junções Aderentes/patologia , Animais , Citrobacter rodentium , Infecções por Enterobacteriaceae/enzimologia , Infecções por Enterobacteriaceae/imunologia , Ativação Enzimática , Feminino , Imunofluorescência , Glucosiltransferases/genética , Glucosiltransferases/imunologia , Mucosa Intestinal/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Mutação , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/imunologia , Fatores de Virulência/genética , Proteínas rho de Ligação ao GTP/metabolismo
2.
Am J Pathol ; 174(2): 436-48, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19147824

RESUMO

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.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Mucosa Intestinal/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Transdução de Sinais/fisiologia , Animais , Western Blotting , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Colágeno , Combinação de Medicamentos , Imunofluorescência , Humanos , Imuno-Histoquímica , Laminina , Microscopia Confocal , Proteoglicanas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Cicatrização/fisiologia
3.
J Immunol ; 181(7): 5035-44, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18802107

RESUMO

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.


Assuntos
Anexina A1/fisiologia , Mediadores da Inflamação/fisiologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Cicatrização/imunologia , Animais , Anexina A1/biossíntese , Anexina A1/deficiência , Anexina A1/genética , Colite/induzido quimicamente , Colite/metabolismo , Colite/patologia , Sulfato de Dextrana/toxicidade , Feminino , Predisposição Genética para Doença , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Índice de Gravidade de Doença , Cicatrização/efeitos dos fármacos
4.
Gastroenterology ; 134(1): 120-30, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18054006

RESUMO

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.


Assuntos
Transformação Celular Neoplásica/patologia , Neoplasias Colorretais/etiologia , Células Epiteliais/fisiologia , Mucosa Intestinal/patologia , Fatores de Transcrição Kruppel-Like/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas ras/fisiologia , Animais , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/patologia , Humanos , Camundongos , Proteínas Proto-Oncogênicas p21(ras)
5.
Mol Biol Cell ; 16(6): 2694-703, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15800062

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/metabolismo , Quimiotaxia de Leucócito , Células Epiteliais/metabolismo , Neutrófilos/fisiologia , Receptores Virais/metabolismo , Junções Íntimas/metabolismo , Anticorpos Monoclonais/metabolismo , Adesão Celular , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/genética , Linhagem Celular , Colo/citologia , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Corantes Fluorescentes , Glutationa Transferase/metabolismo , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Moléculas de Adesão Juncional , Modelos Biológicos , Neutrófilos/química , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Solubilidade
6.
Mol Biol Cell ; 15(8): 3926-37, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15194813

RESUMO

Neutrophil (PMN) transepithelial migration is dependent on the leukocyte beta(2) integrin CD11b/CD18, yet the identity of epithelial counterreceptors remain elusive. Recently, a JAM protein family member termed JAM-C was implicated in leukocyte adhesive interactions; however, its expression in epithelia and role in PMN-epithelial interactions are unknown. Here, we demonstrate that JAM-C is abundantly expressed basolaterally in intestinal epithelia and localizes to desmosomes but not tight junctions. Desmosomal localization of JAM-C was further confirmed by experiments aimed at selective disruption of tight junctions and desmosomes. In assays of PMN transepithelial migration, both JAM-C mAbs and JAM-C/Fc chimeras significantly inhibited the rate of PMN transmigration. Additional experiments revealed specific binding of JAM-C to CD11b/CD18 and provided evidence of other epithelial ligands for CD11b/CD18. These findings represent the first demonstration of direct adhesive interactions between PMN and epithelial intercellular junctions (desmosomes) that regulate PMN transepithelial migration and also suggest that JAM-C may play a role in desmosomal structure/function.


Assuntos
Antígeno CD11b/metabolismo , Antígenos CD18/metabolismo , Moléculas de Adesão Celular/análise , Moléculas de Adesão Celular/metabolismo , Movimento Celular/fisiologia , Desmossomos/química , Imunoglobulinas/análise , Imunoglobulinas/metabolismo , Proteínas de Membrana/análise , Proteínas de Membrana/metabolismo , Neutrófilos/fisiologia , Acrilamida/farmacologia , Moléculas de Adesão Celular/farmacologia , Movimento Celular/efeitos dos fármacos , Desmossomos/efeitos dos fármacos , Desmossomos/metabolismo , Ácido Egtázico/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Humanos , Imunoglobulinas/farmacologia , Filamentos Intermediários/efeitos dos fármacos , Mucosa Intestinal/imunologia , Proteínas de Membrana/farmacologia , Neutrófilos/efeitos dos fármacos
7.
Int J Pharm ; 315(1-2): 122-33, 2006 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-16564652

RESUMO

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.


Assuntos
Eletroporação/métodos , Mucosa Intestinal/citologia , Plasmídeos/genética , RNA Interferente Pequeno/genética , Transfecção/métodos , Linhagem Celular , Eletroporação/normas , Células Epiteliais/metabolismo , Inativação Gênica , Genes Reporter , Humanos , Proteínas/antagonistas & inibidores , Proteínas/genética , Transfecção/normas
8.
Oncogene ; 23(2): 395-402, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14724568

RESUMO

Krüppel-like factor 4 (KLF4 or GKLF) is an inhibitor of the cell cycle. The gene encoding KLF4 is localized on chromosome 9q, previously shown to exhibit allelic loss in colorectal cancer (CRC). In this study, we show that the mean level of KLF4 mRNA in a panel of 30 CRC was 52% that of paired normal colonic tissues. Similarly, the levels of KLF4 mRNA and protein in a panel of six established CRC cell lines were significantly lower than those of an untransformed colonic epithelial cell line. Using highly polymorphic DNA markers that flank the KLF4 locus, we found evidence for loss of heterozygosity (LOH) in two of eight surgically resected CRC specimens. In addition, LOH was observed in five of six CRC cell lines with one additional cell line exhibiting hemizygous deletion in the KLF4 gene. We also found that the 5'-untranslated region of KLF4 was hypermethylated in a subset of resected CRC specimens and cell lines. Lastly, the open-reading frame of KLF4 in two of three CRC cell lines examined contained several point mutations that resulted in a diminished ability to activate the p21(WAF1/Cip1) promoter. These findings indicate that KLF4 is a potential tumor suppressor gene in CRC.


Assuntos
Neoplasias Colorretais/genética , Proteínas de Ligação a DNA/genética , Genes Supressores de Tumor/fisiologia , Fatores de Transcrição/genética , Regiões 5' não Traduzidas/genética , Sequência de Aminoácidos , Sequência de Bases , Southern Blotting , Western Blotting , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like , Perda de Heterozigosidade/genética , Dados de Sequência Molecular , Mutação Puntual/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo
9.
J Cell Sci ; 121(11): 1803-14, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18460584

RESUMO

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.


Assuntos
Citoesqueleto de Actina/metabolismo , Forma Celular/fisiologia , Células Epiteliais/metabolismo , Mucosa Intestinal/embriologia , Mucosa Intestinal/metabolismo , Miosina Tipo II/metabolismo , Actinas/metabolismo , Células CACO-2 , Polaridade Celular/fisiologia , Forma Celular/efeitos dos fármacos , Cofilina 1/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Mucosa Intestinal/citologia , Miosina Tipo II/genética , Nocodazol/farmacologia , Técnicas de Cultura de Órgãos , Polímeros/metabolismo , Estabilidade de RNA , Fosfolipases Tipo C/metabolismo
10.
PLoS One ; 2(7): e658, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17668046

RESUMO

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.


Assuntos
Células Epiteliais/química , Junções Intercelulares/química , Cadeias Pesadas de Miosina/análise , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Linhagem Celular Tumoral/efeitos dos fármacos , Polaridade Celular , Primers do DNA , Regulação para Baixo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Inativação Gênica , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Junções Intercelulares/efeitos dos fármacos , Mucosa Intestinal/química , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos , Cadeias Pesadas de Miosina/genética , Reação em Cadeia da Polimerase , Isoformas de Proteínas/química , RNA Interferente Pequeno/genética
11.
Am J Pathol ; 170(3): 951-66, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17322380

RESUMO

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.


Assuntos
Anexina A2/metabolismo , Movimento Celular/fisiologia , Mucosa Intestinal/metabolismo , Transdução de Sinais/fisiologia , Cicatrização/fisiologia , Proteína rhoA de Ligação ao GTP/metabolismo , Actinas/metabolismo , Células CACO-2 , Células Epiteliais/metabolismo , Imunofluorescência , Humanos , Processamento de Imagem Assistida por Computador , Immunoblotting , Imunoprecipitação , Microscopia Confocal , Análise de Sequência com Séries de Oligonucleotídeos , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
12.
J Immunol ; 179(12): 8112-21, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18056353

RESUMO

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.


Assuntos
Mucosa Intestinal/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptores de Formil Peptídeo/fisiologia , Cicatrização , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/lesões , Inibidores de Fosfoinositídeo-3 Quinase , Receptores de Formil Peptídeo/agonistas , Receptores de Formil Peptídeo/análise
13.
Lab Invest ; 87(10): 1042-54, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17660846

RESUMO

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.


Assuntos
Colite Ulcerativa/microbiologia , Doença de Crohn/microbiologia , Citocinas/metabolismo , Escherichia coli/patogenicidade , Mucosa Intestinal/microbiologia , Células CACO-2 , Estudos de Casos e Controles , Colite Ulcerativa/metabolismo , Doença de Crohn/metabolismo , Escherichia coli/genética , Humanos , Mucosa Intestinal/metabolismo , Permeabilidade
14.
J Exp Med ; 204(13): 3067-76, 2007 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-18039951

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/fisiologia , Colo/metabolismo , Mucosa Intestinal/metabolismo , Receptores de Superfície Celular/fisiologia , Animais , Linhagem Celular Tumoral , Epitélio/embriologia , Predisposição Genética para Doença , Humanos , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Neutrófilos/metabolismo , Permeabilidade
15.
Clin Gastroenterol Hepatol ; 4(10): 1193-8, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16979950

RESUMO

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.


Assuntos
Doença Celíaca/patologia , Intestino Delgado/patologia , Adulto , Biópsia , Diagnóstico Diferencial , Feminino , Humanos
16.
J Biol Chem ; 281(28): 19588-99, 2006 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-16675446

RESUMO

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.


Assuntos
Anexina A1/fisiologia , Células Epiteliais/metabolismo , Receptores de Formil Peptídeo/química , Anexina A1/metabolismo , Anexina A1/farmacologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Movimento Celular , Humanos , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Invasividade Neoplásica , Peptídeos/farmacologia , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/química , Transdução de Sinais , Fatores de Tempo , Cicatrização
17.
J Biol Chem ; 280(12): 11665-74, 2005 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-15677455

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/fisiologia , Integrina beta1/fisiologia , Receptores de Superfície Celular/fisiologia , Proteínas rap1 de Ligação ao GTP/fisiologia , Adesão Celular , Moléculas de Adesão Celular/genética , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Permeabilidade , RNA Interferente Pequeno/farmacologia , Receptores de Superfície Celular/genética
18.
Am J Physiol Gastrointest Liver Physiol ; 287(4): G886-98, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15191880

RESUMO

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.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fator de Crescimento de Hepatócito/farmacologia , Fosfoproteínas/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Actinas/metabolismo , Células CACO-2 , Membrana Celular/metabolismo , Movimento Celular/fisiologia , Células Cultivadas , Colo/citologia , Citosol/metabolismo , Células Epiteliais/citologia , Adesões Focais/efeitos dos fármacos , Adesões Focais/fisiologia , Humanos , Integrina beta1/metabolismo , Paxilina , Fosforilação/efeitos dos fármacos , Regulação para Cima , Proteína rhoA de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA