Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Am J Respir Cell Mol Biol ; 54(4): 515-23, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26393683

RESUMO

MUC1 (MUC in human; Muc in animals) is a transmembrane mucin glycoprotein expressed in mucosal epithelial cells and hematopoietic cells. MUC1 is involved in the resolution of inflammation during airway Pseudomonas aeruginosa (Pa) infection by suppressing Toll-like receptor signaling in airway epithelial cells. Although alveolar macrophages are recognized as critical mediators of cell-mediated immunity against microorganisms inhaled into the airways, the role of MUC1 in regulating their response is unknown. The aims of this study were to determine whether macrophages express MUC1, and, if so, whether MUC1 expression might be associated with macrophage M0/M1/M2 differentiation or phagocytic activity. Human and mouse MUC1/Muc1 expression was drastically up-regulated in classically activated (M1) macrophages compared with nonactivated (M0) and alternatively activated (M2) macrophages. M1 polarization and Pa stimulation each increased MUC1 ectodomain shedding from the macrophage surface in a TNF-α-converting enzyme-dependent manner. MUC1/Muc1 deficiency in M0 macrophages increased adhesion and phagocytosis of Pa and Escherichia coli compared with MUC1/Muc1-expressing cells, and attenuation of phagocytosis by MUC1 was augmented after polarization into M1 macrophages compared with M0 macrophages. Finally, MUC1/Muc1 deficiency in macrophages increased reactive oxygen species production and TNF-α release in response to Pa compared with MUC1/Muc1-sufficient cells. These results indicate that MUC1/Muc1 expression by macrophages is predominantly in the M1 subtype, and that MUC1/Muc1 expression in these cells decreases their phagocytic activity in an antiinflammatory manner.


Assuntos
Macrófagos/imunologia , Mucina-1/fisiologia , Fagocitose/fisiologia , Animais , Aderência Bacteriana , Ensaio de Imunoadsorção Enzimática , Humanos , Macrófagos/metabolismo , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
2.
J Biol Chem ; 290(30): 18316-31, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-25963144

RESUMO

Airway epithelia express sialylated receptors that recognize exogenous danger signals. Regulation of receptor responsiveness to these signals remains incompletely defined. Here, we explore the mechanisms through which the human sialidase, neuraminidase-1 (NEU1), promotes the interaction between the sialoprotein, mucin 1 (MUC1), and the opportunistic pathogen, Pseudomonas aeruginosa. P. aeruginosa flagellin engaged the MUC1 ectodomain (ED), increasing NEU1 association with MUC1. The flagellin stimulus increased the association of MUC1-ED with both NEU1 and its chaperone/transport protein, protective protein/cathepsin A. Scatchard analysis demonstrated NEU1-dependent increased binding affinity of flagellin to MUC1-expressing epithelia. NEU1-driven MUC1-ED desialylation rapidly increased P. aeruginosa adhesion to and invasion of the airway epithelium. MUC1-ED desialylation also increased its shedding, and the shed MUC1-ED competitively blocked P. aeruginosa adhesion to cell-associated MUC1-ED. Levels of desialylated MUC1-ED were elevated in the bronchoalveolar lavage fluid of mechanically ventilated patients with P. aeruginosa airway colonization. Preincubation of P. aeruginosa with these same ex vivo fluids competitively inhibited bacterial adhesion to airway epithelia, and MUC1-ED immunodepletion completely abrogated their inhibitory activity. These data indicate that a prokaryote, P. aeruginosa, in a ligand-specific manner, mobilizes eukaryotic NEU1 to enhance bacterial pathogenicity, but the host retaliates by releasing MUC1-ED into the airway lumen as a hyperadhesive decoy receptor.


Assuntos
Flagelina/metabolismo , Pulmão/metabolismo , Mucina-1/metabolismo , Neuraminidase/metabolismo , Pseudomonas aeruginosa/metabolismo , Adesinas Bacterianas/metabolismo , Aderência Bacteriana , Líquido da Lavagem Broncoalveolar , Linhagem Celular , Humanos , Pulmão/microbiologia , Pulmão/patologia , Ácido N-Acetilneuramínico/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Pseudomonas aeruginosa/patogenicidade
3.
Biochem Biophys Res Commun ; 445(1): 145-50, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24491543

RESUMO

Helicobacter pylori infection of the stomach is associated with the development of gastritis, peptic ulcers, and gastric adenocarcinomas, but the mechanisms are unknown. MUC1 is aberrantly overexpressed by more than 50% of stomach cancers, but its role in carcinogenesis remains to be defined. The current studies were undertaken to identify the genetic mechanisms regulating H. pylori-dependent MUC1 expression by gastric epithelial cells. Treatment of AGS cells with H. pylori increased MUC1 mRNA and protein levels, and augmented MUC1 gene promoter activity, compared with untreated cells. H. pylori increased binding of STAT3 and MUC1 itself to the MUC1 gene promoter within a region containing a STAT3 binding site, and decreased CpG methylation of the MUC1 promoter proximal to the STAT3 binding site, compared with untreated cells. These results suggest that H. pylori upregulates MUC1 expression in gastric cancer cells through STAT3 and CpG hypomethylation.


Assuntos
Regulação da Expressão Gênica , Helicobacter pylori/fisiologia , Interações Hospedeiro-Patógeno , Mucina-1/genética , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Sítios de Ligação/genética , Linhagem Celular Tumoral , Ilhas de CpG/genética , Metilação de DNA , Decitabina , Inibidores Enzimáticos/farmacologia , Humanos , Immunoblotting , Mucina-1/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/microbiologia , Regulação para Cima/efeitos dos fármacos
4.
J Immunol ; 188(4): 2014-22, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22250084

RESUMO

MUC1 is a membrane-tethered mucin glycoprotein expressed on the apical surface of mucosal epithelial cells. Previous in vivo and in vitro studies established that MUC1 counterregulates airway inflammation by suppressing TLR signaling. In this article, we elucidate the mechanism by which MUC1 inhibits TLR5 signaling. Overexpression of MUC1 in HEK293 cells dramatically reduced Pseudomonas aeruginosa-stimulated IL-8 expression and decreased the activation of NF-κB and MAPK compared with cells not expressing MUC1. However, overexpression of MUC1 in HEK293 cells did not affect NF-κB or MAPK activation in response to TNF-α. Overexpression of MyD88 abrogated the ability of MUC1 to inhibit NF-κB activation, and MUC1 overexpression inhibited flagellin-induced association of TLR5/MyD88 compared with controls. The MUC1 cytoplasmic tail associated with TLR5 in all cells tested, including HEK293T cells, human lung adenocarcinoma cell line A549 cells, and human and mouse primary airway epithelial cells. Activation of epidermal growth factor receptor tyrosine kinase with TGF-α induced phosphorylation of the MUC1 cytoplasmic tail at the Y46EKV sequence and increased association of MUC1/TLR5. Finally, in vivo experiments demonstrated increased immunofluorescence colocalization of Muc1/TLR5 and Muc1/phosphotyrosine staining patterns in mouse airway epithelium and increased Muc1 tyrosine phosphorylation in mouse lung homogenates following P. aeruginosa infection. In conclusion, epidermal growth factor receptor tyrosine phosphorylates MUC1, leading to an increase in its association with TLR5, thereby competitively and reversibly inhibiting recruitment of MyD88 to TLR5 and downstream signaling events. This unique ability of MUC1 to control TLR5 signaling suggests its potential role in the pathogenesis of chronic inflammatory lung diseases.


Assuntos
Receptores ErbB/metabolismo , Mucina-1/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Mucosa Respiratória/metabolismo , Receptor 5 Toll-Like/metabolismo , Animais , Linhagem Celular , Células Epiteliais/metabolismo , Receptores ErbB/biossíntese , Flagelina/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Interleucina-8/biossíntese , Pulmão/metabolismo , Pneumopatias/imunologia , Pneumopatias/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/biossíntese , Proteínas Quinases Ativadas por Mitógeno/imunologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mucina-1/biossíntese , Fator 88 de Diferenciação Mieloide/biossíntese , NF-kappa B/biossíntese , NF-kappa B/metabolismo , Fosforilação , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/imunologia , Pseudomonas aeruginosa/patogenicidade , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
5.
Am J Physiol Gastrointest Liver Physiol ; 303(6): G765-74, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22766852

RESUMO

MUC1 is a membrane-tethered mucin expressed on the apical surface of epithelial cells. Our previous report (Guang W, Ding H, Czinn SJ, Kim KC, Blanchard TG, Lillehoj EP. J Biol Chem 285: 20547-20557, 2010) demonstrated that expression of MUC1 in AGS gastric epithelial cells limits Helicobacter pylori infection and reduces bacterial-driven IL-8 production. In this study, we identified the peroxisome proliferator-associated receptor-γ (PPARγ) upstream of MUC1 in the anti-inflammatory pathway suppressing H. pylori- and phorbol 12-myristate 13-acetate (PMA)-stimulated IL-8 production. Treatment of AGS cells with H. pylori or PMA increased IL-8 levels in cell culture supernatants compared with cells treated with the respective vehicle controls. Prior small interfering (si)RNA-induced MUC1 silencing further increased H. pylori- and PMA-stimulated IL-8 levels compared with a negative control siRNA. MUC1-expressing AGS cells pretreated with the PPARγ agonist troglitazone (TGN) had reduced H. pylori- and PMA-stimulated IL-8 levels compared with cells treated with H. pylori or PMA alone. However, following MUC1 siRNA knockdown, no differences in IL-8 levels were seen between TGN/H. pylori and H. pylori-only cells or between TGN/PMA and PMA-only cells. Finally, TGN-treated AGS cells had increased Muc1 promoter activity, as measured using a Muc1-luciferase reporter gene, and greater MUC1 protein levels by Western blot analysis, compared with vehicle controls. These results support the hypothesis that PPARγ stimulates MUC1 expression by AGS cells, thereby attenuating H. pylori- and PMA-induced IL-8 production.


Assuntos
Células Epiteliais/metabolismo , Interleucina-8/metabolismo , PPAR gama/metabolismo , Estômago/citologia , Cromanos/farmacologia , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Deleção de Genes , Regulação da Expressão Gênica/fisiologia , Helicobacter pylori , Humanos , Interleucina-8/genética , PPAR gama/genética , Dibutirato de 12,13-Forbol , Regiões Promotoras Genéticas , Tiazolidinedionas/farmacologia , Troglitazona
6.
J Biol Chem ; 285(27): 20547-57, 2010 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-20430889

RESUMO

Helicobacter pylori infection of the gastric mucosa causes an active-chronic inflammation that is strongly linked to the development of duodenal and gastric ulcers and stomach cancer. However, greater than 80% of individuals infected with H. pylori are asymptomatic beyond histologic inflammation, and it is unknown what factors influence the incidence and character of bacterial-associated gastritis and related disorders. Because previous studies demonstrated that the Muc1 epithelial glycoprotein inhibited inflammation during acute lung infection by Pseudomonas aeruginosa, we asked whether Muc1 might also counter-regulate gastric inflammation in response to H. pylori infection. Muc1(-/-) mice displayed increased bacterial colonization of the stomach and greater TNF-alpha and keratinocyte chemoattractant transcript levels compared with Muc1(+/+) mice after experimental H. pylori infection. Knockdown of Muc1 expression in AGS human gastric epithelial cells by RNA interference was associated with increased phosphorylation of IkappaBalpha, augmented activation and nuclear translocation of NF-kappaB, and enhanced production of interleulin-8 compared with Muc1-expressing cells. Conversely, Muc1 overexpression was correlated with decreased NF-kappaB activation, reduced interleulin-8 production, and diminished IkappaB kinase beta (IKKbeta)/IKKgamma coimmunoprecipitation compared with cells expressing Muc1 endogenously. Cotransfection of AGS cells with Muc1 plus IKKbeta, but not a catalytically inactive IKKbeta mutant, reversed the Muc1 inhibitory effect. Finally, Muc1 formed a coimmunoprecipitation complex with IKKgamma but not with IKKbeta. These results are consistent with the hypothesis that Muc1 binds to IKKgamma, thereby inhibiting formation of the catalytically active IKK complex and blocking the ability of H. pylori to stimulate IkappaBalpha phosphorylation, NF-kappaB activation, and downstream inflammatory responses.


Assuntos
Técnicas de Silenciamento de Genes , Mucina-1/genética , Animais , Toxina da Cólera/farmacologia , Ensaio de Unidades Formadoras de Colônias , DNA/genética , DNA/isolamento & purificação , Ensaio de Imunoadsorção Enzimática , Feminino , Mucosa Gástrica/metabolismo , Amplificação de Genes , Regulação da Expressão Gênica , Genes Reporter , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/patologia , Helicobacter pylori , Quinase I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Interleucina-8/metabolismo , Masculino , Camundongos , Camundongos Knockout , Inibidor de NF-kappaB alfa , Recombinação Genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estômago/patologia , Transfecção , Fator de Necrose Tumoral alfa/genética
7.
Proteome Sci ; 9: 4, 2011 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-21251289

RESUMO

BACKGROUND: Airway surface liquid, often referred to as mucus, is a thin layer of fluid covering the luminal surface that plays an important defensive role against foreign particles and chemicals entering the lungs. Airway mucus contains various macromolecules, the most abundant being mucin glycoproteins, which contribute to its defensive function. Airway epithelial cells cultured in vitro secrete mucins and nonmucin proteins from their apical surface that mimics mucus production in vivo. The current study was undertaken to identify the polypeptide constituents of human airway epithelial cell secretions to gain a better understanding of the protein composition of respiratory mucus. RESULTS: Fifty-five proteins were identified in the high molecular weight fraction of apical secretions collected from in vitro cultures of well-differentiated primary human airway epithelial cells and isolated under physiological conditions. Among these were MUC1, MUC4, MUC5B, and MUC16 mucins. By proteomic analysis, the nonmucin proteins could be classified as inflammatory, anti-inflammatory, anti-oxidative, and/or anti-microbial. CONCLUSIONS: Because the majority of the nonmucin proteins possess molecular weights less than that selected for analysis, it is theoretically possible that they may associate with the high molecular weight and negatively charged mucins to form a highly ordered structural organization that is likely to be important for maintaining the proper defensive function of airway mucus.

8.
Exp Lung Res ; 37(10): 606-14, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22044398

RESUMO

Guaifenesin is widely used to alleviate symptoms of excessive mucus accumulation in the respiratory tract. However, its mechanism of action is poorly understood. The authors hypothesized that guaifenesin improves mucociliary clearance in humans by reducing mucin release, by decreasing mucus viscoelasticity, and by increasing mucociliary transport. To test these hypotheses, human differentiated airway epithelial cells, cultured at an air-liquid interface, were treated with clinically relevant concentrations of guaifenesin by addition to the basolateral medium. To evaluate the effect on mucin secretion, the authors used an anzyme-linked immunosorbent assay (ELISA) to measure the amounts of MUC5AC protein in apical surface fluid and cell lysates. To measure mucociliary transportability, additional cultures were treated for 1 or 6 hours with guaifenesin, and the movement of cell debris was measured from video data. Further, the authors measured mucus dynamic viscoelasticity using a micro cone and plate rheometer with nondestructive creep transformation. Guaifenesin suppressed mucin production in a dose-dependent manner at clinically relevant concentrations. The reduced mucin production was associated with increased mucociliary transport and decreased viscoelasticity of the mucus. Viability of the cultures was not significantly affected. These results suggest that guaifenesin could improve mucociliary clearance in humans by reducing the release and/or production of mucins, thereby altering mucus rheology.


Assuntos
Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Guaifenesina/farmacologia , Mucinas/biossíntese , Depuração Mucociliar/efeitos dos fármacos , Brônquios/citologia , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Células Epiteliais/citologia , Humanos , Mucina-5AC/metabolismo , Mucinas/metabolismo , Muco/efeitos dos fármacos , Muco/metabolismo , Reologia/métodos , Traqueia/citologia , Traqueia/efeitos dos fármacos , Traqueia/metabolismo , Viscosidade/efeitos dos fármacos
9.
Am J Physiol Lung Cell Mol Physiol ; 298(4): L558-63, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20081068

RESUMO

MUC1 is a transmembrane glycoprotein expressed on the apical surface of airway epithelial cells and plays an anti-inflammatory role during airway bacterial infection. In this study, we determined whether the anti-inflammatory effect of MUC1 is also operative during the respiratory syncytial virus (RSV) infection. The lung epithelial cell line A549 was treated with RSV, and the production of TNFalpha and the levels of MUC1 protein were monitored temporally during the course of infection by ELISA and Western blot analysis. Small inhibitory RNA (siRNA) transfection was utilized to assess the role of MUC1 in regulating RSV-mediated inflammatory responses by lung epithelial cells. Our results revealed that: 1) following RSV infection, an increase in MUC1 level was preceded by an increase in TNFalpha production and completely inhibited by soluble TNF receptor (TNFR); and 2) knockdown of MUC1 using MUC1 siRNA resulted in a greater increase in TNFalpha level following RSV infection compared with control siRNA treatment. We conclude that the RSV-induced increase in the TNFalpha levels upregulates MUC1 through its interaction with TNFR, which in turn suppresses further increase in TNFalpha by RSV, thus forming a negative feedback loop in the control of RSV-induced inflammation. This is the first demonstration showing that MUC1 can suppress the virus-induced inflammatory responses.


Assuntos
Anti-Inflamatórios/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Pulmão/patologia , Mucina-1/metabolismo , Infecções por Vírus Respiratório Sincicial/metabolismo , Vírus Sinciciais Respiratórios/fisiologia , Animais , Linhagem Celular Tumoral , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Pulmão/virologia , Camundongos , Mucina-1/biossíntese , Mucina-1/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Infecções por Vírus Respiratório Sincicial/genética , Solubilidade , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
10.
Am J Respir Cell Mol Biol ; 39(6): 644-7, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18617677

RESUMO

MUC1 is a membrane-tethered mucin expressed on the surface of epithelial cells lining mucosal surfaces. Recent studies have begun to elucidate the physiologic function of MUC1 in the airways, pointing to an antiinflammatory role that is initiated late in the course of bacterial infection and is mediated through inhibition of TLR signaling. These new findings have great potential for clinical applications in controlling excessive and prolonged lung inflammation. This review briefly summarizes the protein structural features of MUC1 relevant to its function, the discovery of its antiinflammatory properties, and potential directions for future avenues of study.


Assuntos
Pulmão/metabolismo , Mucina-1/metabolismo , Animais , Anti-Inflamatórios/metabolismo , Humanos , Inflamação , Pulmão/patologia , Mucina-1/química
11.
Am J Respir Cell Mol Biol ; 38(3): 263-8, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18079492

RESUMO

MUC1 (MUC1 in humans and Muc1 in nonhuman species) is a transmembrane mucin-like glycoprotein expressed in epithelial cells lining various mucosal surfaces as well as hematopoietic cells. Recently, we showed that Muc1(-/-) mice exhibited greater inflammatory responses to Pseudomonas aeruginosa or its flagellin compared with their wild-type littermates, and our studies with cultured cells revealed that MUC1/Muc1 suppressed the Toll-like receptor (TLR) 5 signaling pathway, suggesting its anti-inflammatory role. Here we demonstrate that other TLR signaling (TLR2, 3, 4, 7, and 9) is also suppressed by MUC1/Muc1. The results from this study suggest that MUC1/Muc1 may play a crucial role during airway infection and inflammation by various pathogenic bacteria and viruses.


Assuntos
Mucina-1/genética , Mucina-1/metabolismo , Transdução de Sinais , Receptores Toll-Like/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Quimiocina CXCL1/análise , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Humanos , Rim/citologia , Luciferases de Renilla/metabolismo , Macrófagos Alveolares/citologia , Macrófagos Alveolares/metabolismo , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Knockout , Proteínas Recombinantes/metabolismo , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo , Estatística como Assunto , Traqueia/citologia , Transfecção , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
12.
Biochim Biophys Acta ; 1773(7): 1028-38, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17524503

RESUMO

beta-Catenin binds to the cytoplasmic region of the type 1 membrane glycoprotein MUC1. In the current study, we utilized HEK293T cells expressing the full-length MUC1 protein, or a CD8/MUC1 fusion protein containing only the MUC1 cytoplasmic tail, to investigate the effects of beta-catenin binding to MUC1 on downstream beta-catenin-dependent events. Compared with HEK293T cells transfected with empty vector or CD8 alone, expression of the MUC1 cytoplasmic tail inhibited beta-catenin binding to E-cadherin, decreased translocation of beta-catenin into the nucleus, reduced activation of the LEF-1 transcription factor, and blocked expression of the cyclin D1 and c-Myc proteins. Furthermore, expression of MUC1 was associated with decreased cell proliferation, either in the context of the transfected HEK293T cells, or when comparing wild type (Muc1(+/+)) vs. knockout (Muc1(-/-)) mouse primary tracheal epithelial cells. We conclude that MUC1 inhibits cell proliferation through a beta-catenin/LEF-1/cyclin D1/c-Myc pathway.


Assuntos
Antígenos de Neoplasias/metabolismo , Proliferação de Células , Mucinas/metabolismo , beta Catenina/metabolismo , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias/genética , Antígenos CD8/genética , Antígenos CD8/metabolismo , Caderinas/metabolismo , Células Cultivadas , Ciclina D1/metabolismo , Humanos , Fator 1 de Ligação ao Facilitador Linfoide/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Camundongos , Dados de Sequência Molecular , Mucina-1 , Mucinas/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Transcrição Gênica
13.
Am J Respir Cell Mol Biol ; 37(6): 691-8, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17600314

RESUMO

We previously reported that neutrophil elastase (NE) stimulated MUC1 gene expression in A549 lung epithelial cells through binding of Sp1 to the MUC1 promoter element. The current study was undertaken to elucidate the complete signaling pathway leading to Sp1 activation. Using a combination of pharmacologic inhibitors, dominant-negative mutant, RNA interference, and soluble receptor blocking techniques, we identified a protein kinase Cdelta (PKCdelta) --> dual oxidase 1 (Duox1) --> reactive oxygen species (ROS) --> TNF-alpha-converting enzyme (TACE) --> TNF-alpha --> TNF receptor (TNFR)1 --> extracellular signal-regulated kinase (ERK)1/2 --> Sp1 pathway as responsible for NE-activated MUC1 transcription. This cascade was identical up to the point of TACE with the signaling pathway previously reported for NE-stimulated MUC5AC production. However, unlike the MUC5AC pathway, TNF-alpha, TNFR1, ERK1/2, and Sp1 were unique components of the MUC1 pathway. Given the anti-inflammatory role of MUC1 during airway bacterial infection, up-regulation of MUC1 by inflammatory mediators such as NE and TNF-alpha suggests a crucial role for MUC1 in the control of excessive inflammation during airway bacterial infection.


Assuntos
Elastase de Leucócito/metabolismo , Mucina-1/genética , Transdução de Sinais , Transcrição Gênica , Proteínas ADAM/metabolismo , Proteína ADAM17 , Linhagem Celular Tumoral , Células Cultivadas , Oxidases Duais , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavoproteínas/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Modelos Biológicos , NADPH Oxidases/antagonistas & inibidores , NF-kappa B/metabolismo , Fosfotreonina/metabolismo , Proteína Quinase C-delta/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Transcrição Sp1/metabolismo , Transcrição Gênica/efeitos dos fármacos
14.
Int J Oncol ; 26(5): 1169-76, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15809706

RESUMO

Apoptosis is a physiological mechanism responsible for a wide range of cellular processes during growth, development, carcinogenesis, and inflammation. In this study, we determined whether MUC1 affects Fas ligand (FasL)-induced apoptosis using MUC1 expressing (MUC1(+)) and MUC1(-) cells. Following treatment with 50 nM FasL, apoptosis, caspase-8 activity, and cell surface Fas receptor were measured by cytosolic nucleosome ELISA, colorimetric enzyme assay, and immunofluorescence analysis, respectively. Our results showed that (i) treatment with FasL increased caspase-8 activity (maximum at 4 h) and apoptosis (maximum at 8 h) in both MUC1(+) and MUC1(-) cells, (ii) FasL-induced caspase-8 activity and apoptosis were significantly greater in MUC1(+) cells compared with MUC1(-) cells, (iii) FasL treatment increased cell surface expression of Fas receptor in MUC1(+) cells to a greater extent compared with MUC1(-) cells, (iv) increased cell surface expression of Fas in MUC1(+) cells was not blocked by an inhibitor of protein synthesis (cycloheximide), but was completely abrogated by brefeldin A, an inhibitor of post-translational protein trafficking to the cell surface, and (v) brefeldin A inhibited the increased sensitivity of MUC1(+) cells to FasL-induced apoptosis. We conclude that MUC1(+) cells were more sensitive to FasL-induced apoptosis compared with MUC1(-) cells due to up-regulation of Fas receptor on the cell surface by a mechanism involving increased intracellular trafficking.


Assuntos
Apoptose , Glicoproteínas de Membrana/metabolismo , Mucina-1/metabolismo , Animais , Antígenos de Superfície , Células CHO , Caspase 8 , Caspases/metabolismo , Técnicas de Cultura de Células , Membrana Celular/fisiologia , Cricetinae , Cricetulus , Ensaio de Imunoadsorção Enzimática , Proteína Ligante Fas , Imunofluorescência , Ligantes , Regulação para Cima , Receptor fas/biossíntese
15.
Anticancer Res ; 24(1): 235-40, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15015602

RESUMO

Muc1 is the cell surface glycoprotein abundantly expressed in cancer cells and has been shown to be involved in tumor metastasis and promotion. Recently, we identified a 37 bp segment on the hamster Muc1 promoter with the ability to suppress Muc1 transcription. This 37 bp putative negative regulatory element (NRE) binds to a transcriptional regulator Yin Yang 1 (YY1). In the present study, we examined whether binding of YY1 is responsible for the negative regulatory effect by the 37 bp segment using a hamster pancreatic cancer cell line, HP-1 cells, transfected with various expression plasmid constructs. Our results showed that: (1) overexpression of YY1 up-regulated the transcriptional activity of the full-length hamster Muc1 promoter in a dose-dependent manner; (2) the mutation of the YY1 binding site did not affect either the basal transcriptional activity or the increased transcriptional activity by YY1; and (3) even the deletion of the 37 bp NRE segment could not abrogate the increased transcriptional activity by YY1. We conclude that the NRE acts in a YY1-independent manner and that YY1 instead enhances Muc1 transcriptional activity. Further study of the precise mechanism by which YY1 augments Muc1 gene expression should be worthwhile.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Mucina-1/genética , Neoplasias Pancreáticas/genética , Fatores de Transcrição/fisiologia , Animais , Linhagem Celular Tumoral , Cricetinae , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fatores de Ligação de DNA Eritroide Específicos , Regulação Neoplásica da Expressão Gênica , Genes Reguladores , Mutação , Neoplasias Pancreáticas/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transfecção , Fator de Transcrição YY1
16.
Arch Pharm Res ; 25(6): 770-80, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12510824

RESUMO

The airway surface liquid (ASL), often referred to as mucus, is a thin layer of fluid covering the luminal surface of the airway. The major function of mucus is to protect the lung through mucociliary clearance against foreign particles and chemicals entering the lung. The mucus is comprised of water, ions, and various kinds of macromolecules some of which possess the protective functions such as anti-microbial, anti-protease, and anti-oxidant activity. Mucus glycoproteins or mucins are mainly responsible for the viscoelastic property of mucus, which is crucial for the effective mucociliary clearance. There are at least eight mucin genes identified in the human airways, which will potentially generate various kinds of mucin molecules. At present, neither the exact structures of mucin proteins nor their regulation are understood although it seems likely that different types of mucins are involved in different functions and might also be associated with certain airway diseases. The fact that mucins are tightly associated with various macromolecules present in ASL seems to suggest that the defensive role of ASL is determined not only by these individual components but rather by a combination of these components. Collectively, mucins in ASL may be compared to aircraft carriers carrying various types of weapons in defense of airbome enemies.


Assuntos
Pulmão/fisiologia , Muco/química , Muco/fisiologia , Animais , Humanos , Mucinas/química , Mucinas/fisiologia , Depuração Mucociliar/fisiologia
17.
Clin Vaccine Immunol ; 17(8): 1196-202, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20592113

RESUMO

Pseudomonas aeruginosa lung infection is a major cause of morbidity and mortality worldwide. P. aeruginosa flagellin, the main structural protein of the flagellar filament, is a virulence factor with proinflammatory activity on respiratory epithelial cells. P. aeruginosa bacteria express one of two isoforms of flagellin (type a or b) that differ in their primary amino acid sequences as well as in posttranslational glycosylation. In this study, the distribution of type a and b flagellins among 3 P. aeruginosa laboratory strains and 14 clinical isolates (1 ulcerative keratitis, 3 cystic fibrosis, and 10 acute pneumonia isolates) was determined, and their abilities to stimulate interleukin-8 (IL-8) production by human airway epithelial cells was compared. By comparison with the PAK (type a) and PAO1 (type b) prototype laboratory strains, 10/14 (71.4%) of clinical isolates expressed type a and 4/14 (28.6%) expressed type b flagellins. Among four cell lines surveyed, BEAS-2B cells were found to give the greatest difference between constitutive and flagellin-stimulated IL-8 production. All 17 flagellins stimulated IL-8 production by BEAS-2B cells (range, 700 to 4,000 pg/ml). However, no discernible differences in IL-8 production were evident when comparing type a versus type b flagellins or flagellins from laboratory versus clinical strains or among the clinical strains.


Assuntos
Células Epiteliais/imunologia , Flagelina/biossíntese , Flagelina/imunologia , Interleucina-8/metabolismo , Pseudomonas aeruginosa/imunologia , Linhagem Celular , Flagelina/classificação , Humanos , Immunoblotting , Isoformas de Proteínas/imunologia , Mucosa Respiratória/imunologia
18.
Int J Biochem Cell Biol ; 41(6): 1354-60, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19135167

RESUMO

MUC1 is an integral membrane glycoprotein expressed on epithelial and hematopoietic cells with a COOH-terminus (CT) that mediates intracellular signal transduction. To better understand MUC1-dependent signaling, we searched for proteins binding to its CT using the yeast two-hybrid system with the MUC1 CT as bait and a human epithelial cell cDNA library as prey. Of the six positive clones identified, all encoded calcium-modulating cyclophilin ligand (CAML). The MUC1 CT interacted with CAML in transformed yeast cells as revealed by growth on selective media and in situ X-alpha-galactosidase activity. Binding of the MUC1 CT to CAML in human epithelial cells was confirmed by reciprocal coimmunoprecipitations, confocal microscopy, protein crosslinking, and coupled transcription/translation analyses. By deletion mutagenesis, the NH(2)-terminus of CAML was responsible for binding to the MUC1 CT. Finally, transfection of cells with plasmids encoding MUC1 and CAML increased intracellular calcium levels compared with cells transfected with either plasmid alone, suggesting a possible biological significance of the MUC1-CAML interaction.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Mucina-1/metabolismo , Western Blotting , Neoplasias da Mama/metabolismo , Cálcio/metabolismo , Linhagem Celular Tumoral , Feminino , Biblioteca Gênica , Humanos , Microscopia Confocal , Mucina-1/genética , Plasmídeos , RNA Mensageiro/genética , Transdução de Sinais , Transfecção , Técnicas do Sistema de Duplo-Híbrido
19.
J Immunol ; 180(11): 7655-63, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18490768

RESUMO

Allergic asthma, an inflammatory disease characterized by the infiltration and activation of various leukocytes, the production of Th2 cytokines and leukotrienes, and atopy, also affects the function of other cell types, causing goblet cell hyperplasia/hypertrophy, increased mucus production/secretion, and airway hyperreactivity. Eosinophilic inflammation is a characteristic feature of human asthma, and recent evidence suggests that eosinophils also play a critical role in T cell trafficking in animal models of asthma. Nicotine is an anti-inflammatory, but the association between smoking and asthma is highly contentious and some report that smoking cessation increases the risk of asthma in ex-smokers. To ascertain the effects of nicotine on allergy/asthma, Brown Norway rats were treated with nicotine and sensitized and challenged with allergens. The results unequivocally show that, even after multiple allergen sensitizations, nicotine dramatically suppresses inflammatory/allergic parameters in the lung including the following: eosinophilic/lymphocytic emigration; mRNA and/or protein expression of the Th2 cytokines/chemokines IL-4, IL-5, IL-13, IL-25, and eotaxin; leukotriene C(4); and total as well as allergen-specific IgE. Although nicotine did not significantly affect hexosaminidase release, IgG, or methacholine-induced airway resistance, it significantly decreased mucus content in bronchoalveolar lavage; interestingly, however, despite the strong suppression of IL-4/IL-13, nicotine significantly increased the intraepithelial-stored mucosubstances and Muc5ac mRNA expression. These results suggest that nicotine modulates allergy/asthma primarily by suppressing eosinophil trafficking and suppressing Th2 cytokine/chemokine responses without reducing goblet cell metaplasia or mucous production and may explain the lower risk of allergic diseases in smokers. To our knowledge this is the first direct evidence that nicotine modulates allergic responses.


Assuntos
Alérgenos/imunologia , Asma/imunologia , Células Caliciformes/imunologia , Pulmão/imunologia , Células Musculares/imunologia , Nicotina/farmacologia , Células Th2/imunologia , Ambrosia/imunologia , Animais , Asma/metabolismo , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Regulação para Baixo , Eosinófilos/imunologia , Eosinófilos/metabolismo , Feminino , Células Caliciformes/metabolismo , Imunoglobulina E/imunologia , Imunoglobulina E/metabolismo , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Leucotrieno C4/biossíntese , Leucotrieno C4/imunologia , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Mucina-5AC , Mucinas/metabolismo , Células Musculares/metabolismo , Ratos , Ratos Endogâmicos BN , Células Th2/efeitos dos fármacos , Células Th2/metabolismo
20.
Am J Physiol Lung Cell Mol Physiol ; 293(3): L686-92, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17586693

RESUMO

MUC1 is a membrane-tethered mucin-like glycoprotein expressed on the surface of various mucosal epithelial cells as well as hematopoietic cells. Recently, we showed that MUC1 suppresses flagellin-induced Toll-like receptor (TLR) 5 signaling both in vivo and in vitro through cross talk with TLR5. In this study, we determined whether phosphoinositide 3-kinase (PI3K), a negative regulator of TLR5 signaling, is involved in the cross talk between MUC1 and TLR5 using various genetically modified epithelial cell lines. Our results showed 1) activation of MUC1 induced recruitment of the PI3K regulatory subunit p85 to the MUC1 cytoplasmic tail (CT) as well as Akt phosphorylation, 2) MUC1-induced Akt phosphorylation required the presence of Tyr(20) within the PI3K binding motif of the MUC1 CT, and 3) mutation of Tyr(20) or pharmacological inhibition of PI3K activation failed to block MUC1-induced suppression of TLR5 signaling. We conclude that whereas PI3K is downstream of MUC1 activation and negatively regulates TLR5 signaling, it is not responsible for MUC1-induced suppression of TLR5 signaling.


Assuntos
Mucinas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Receptor 5 Toll-Like/metabolismo , Anticorpos Monoclonais/farmacologia , Antígenos CD8/metabolismo , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Humanos , Inflamação , Mucinas/química , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA