Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Am J Respir Cell Mol Biol ; 54(4): 515-23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26393683

ABSTRACT

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.


Subject(s)
Macrophages/immunology , Mucin-1/physiology , Phagocytosis/physiology , Animals , Bacterial Adhesion , Enzyme-Linked Immunosorbent Assay , Humans , Macrophages/metabolism , Mice , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
J Biol Chem ; 290(30): 18316-31, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-25963144

ABSTRACT

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.


Subject(s)
Flagellin/metabolism , Lung/metabolism , Mucin-1/metabolism , Neuraminidase/metabolism , Pseudomonas aeruginosa/metabolism , Adhesins, Bacterial/metabolism , Bacterial Adhesion , Bronchoalveolar Lavage Fluid , Cell Line , Humans , Lung/microbiology , Lung/pathology , N-Acetylneuraminic Acid/metabolism , Protein Binding , Protein Structure, Tertiary , Pseudomonas aeruginosa/pathogenicity
3.
Biochem Biophys Res Commun ; 445(1): 145-50, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24491543

ABSTRACT

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.


Subject(s)
Gene Expression Regulation , Helicobacter pylori/physiology , Host-Pathogen Interactions , Mucin-1/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Binding Sites/genetics , Cell Line, Tumor , CpG Islands/genetics , DNA Methylation , Decitabine , Enzyme Inhibitors/pharmacology , Humans , Immunoblotting , Mucin-1/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Up-Regulation/drug effects
4.
Am J Physiol Gastrointest Liver Physiol ; 303(6): G765-74, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22766852

ABSTRACT

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.


Subject(s)
Epithelial Cells/metabolism , Interleukin-8/metabolism , PPAR gamma/metabolism , Stomach/cytology , Chromans/pharmacology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/cytology , Epithelial Cells/drug effects , Gene Deletion , Gene Expression Regulation/physiology , Helicobacter pylori , Humans , Interleukin-8/genetics , PPAR gamma/genetics , Phorbol 12,13-Dibutyrate , Promoter Regions, Genetic , Thiazolidinediones/pharmacology , Troglitazone
5.
J Immunol ; 188(4): 2014-22, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22250084

ABSTRACT

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.


Subject(s)
ErbB Receptors/metabolism , Mucin-1/metabolism , Myeloid Differentiation Factor 88/metabolism , Respiratory Mucosa/metabolism , Toll-Like Receptor 5/metabolism , Animals , Cell Line , Epithelial Cells/metabolism , ErbB Receptors/biosynthesis , Flagellin/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Interleukin-8/biosynthesis , Lung/metabolism , Lung Diseases/immunology , Lung Diseases/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/biosynthesis , Mitogen-Activated Protein Kinases/immunology , Mitogen-Activated Protein Kinases/metabolism , Mucin-1/biosynthesis , Myeloid Differentiation Factor 88/biosynthesis , NF-kappa B/biosynthesis , NF-kappa B/metabolism , Phosphorylation , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/pathogenicity , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
6.
Exp Lung Res ; 37(10): 606-14, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22044398

ABSTRACT

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.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/metabolism , Guaifenesin/pharmacology , Mucins/biosynthesis , Mucociliary Clearance/drug effects , Bronchi/cytology , Bronchi/drug effects , Bronchi/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Epithelial Cells/cytology , Humans , Mucin 5AC/metabolism , Mucins/metabolism , Mucus/drug effects , Mucus/metabolism , Rheology/methods , Trachea/cytology , Trachea/drug effects , Trachea/metabolism , Viscosity/drug effects
7.
Proteome Sci ; 9: 4, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21251289

ABSTRACT

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.
Clin Vaccine Immunol ; 17(8): 1196-202, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20592113

ABSTRACT

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.


Subject(s)
Epithelial Cells/immunology , Flagellin/biosynthesis , Flagellin/immunology , Interleukin-8/metabolism , Pseudomonas aeruginosa/immunology , Cell Line , Flagellin/classification , Humans , Immunoblotting , Protein Isoforms/immunology , Respiratory Mucosa/immunology
9.
J Biol Chem ; 285(27): 20547-57, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20430889

ABSTRACT

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.


Subject(s)
Gene Knockdown Techniques , Mucin-1/genetics , Animals , Cholera Toxin/pharmacology , Colony-Forming Units Assay , DNA/genetics , DNA/isolation & purification , Enzyme-Linked Immunosorbent Assay , Female , Gastric Mucosa/metabolism , Gene Amplification , Gene Expression Regulation , Genes, Reporter , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Helicobacter pylori , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , Interleukin-8/metabolism , Male , Mice , Mice, Knockout , NF-KappaB Inhibitor alpha , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Stomach/pathology , Transfection , Tumor Necrosis Factor-alpha/genetics
10.
Am J Physiol Lung Cell Mol Physiol ; 298(4): L558-63, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20081068

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/metabolism , Epithelial Cells/metabolism , Epithelial Cells/virology , Lung/pathology , Mucin-1/metabolism , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Viruses/physiology , Animals , Cell Line, Tumor , Feedback, Physiological , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Lung/virology , Mice , Mucin-1/biosynthesis , Mucin-1/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Respiratory Syncytial Virus Infections/genetics , Solubility , Time Factors , Tumor Necrosis Factor-alpha/metabolism
11.
Int J Biochem Cell Biol ; 41(6): 1354-60, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19135167

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Mucin-1/metabolism , Blotting, Western , Breast Neoplasms/metabolism , Calcium/metabolism , Cell Line, Tumor , Female , Gene Library , Humans , Microscopy, Confocal , Mucin-1/genetics , Plasmids , RNA, Messenger/genetics , Signal Transduction , Transfection , Two-Hybrid System Techniques
12.
Am J Respir Cell Mol Biol ; 39(6): 644-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18617677

ABSTRACT

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.


Subject(s)
Lung/metabolism , Mucin-1/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Humans , Inflammation , Lung/pathology , Mucin-1/chemistry
13.
J Immunol ; 180(11): 7655-63, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18490768

ABSTRACT

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.


Subject(s)
Allergens/immunology , Asthma/immunology , Goblet Cells/immunology , Lung/immunology , Muscle Cells/immunology , Nicotine/pharmacology , Th2 Cells/immunology , Ambrosia/immunology , Animals , Asthma/metabolism , Bronchoalveolar Lavage Fluid/immunology , Cytokines/immunology , Cytokines/metabolism , Down-Regulation , Eosinophils/immunology , Eosinophils/metabolism , Female , Goblet Cells/metabolism , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Leukotriene C4/biosynthesis , Leukotriene C4/immunology , Lung/cytology , Lung/drug effects , Lung/metabolism , Mucin 5AC , Mucins/metabolism , Muscle Cells/metabolism , Rats , Rats, Inbred BN , Th2 Cells/drug effects , Th2 Cells/metabolism
14.
Am J Respir Cell Mol Biol ; 38(3): 263-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18079492

ABSTRACT

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.


Subject(s)
Mucin-1/genetics , Mucin-1/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , Animals , Cell Line , Cells, Cultured , Chemokine CXCL1/analysis , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Gene Expression Regulation , Genes, Reporter , Humans , Kidney/cytology , Luciferases, Renilla/metabolism , Macrophages, Alveolar/cytology , Macrophages, Alveolar/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Recombinant Proteins/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Statistics as Topic , Trachea/cytology , Transfection , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
Am J Respir Cell Mol Biol ; 37(6): 691-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17600314

ABSTRACT

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.


Subject(s)
Leukocyte Elastase/metabolism , Mucin-1/genetics , Signal Transduction , Transcription, Genetic , ADAM Proteins/metabolism , ADAM17 Protein , Cell Line, Tumor , Cells, Cultured , Dual Oxidases , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavoproteins/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Humans , Models, Biological , NADPH Oxidases/antagonists & inhibitors , NF-kappa B/metabolism , Phosphothreonine/metabolism , Protein Kinase C-delta/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Sp1 Transcription Factor/metabolism , Transcription, Genetic/drug effects
16.
Am J Physiol Lung Cell Mol Physiol ; 293(3): L686-92, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17586693

ABSTRACT

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.


Subject(s)
Mucins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Toll-Like Receptor 5/metabolism , Antibodies, Monoclonal/pharmacology , CD8 Antigens/metabolism , Cell Line , Enzyme Activation/drug effects , Humans , Inflammation , Mucins/chemistry , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Tyrosine/metabolism
17.
Am J Physiol Lung Cell Mol Physiol ; 293(3): L693-701, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17575006

ABSTRACT

The current study was conducted to elucidate the mechanism through which TNF-alpha stimulates expression of MUC1, a membrane-tethered mucin. A549 human lung alveolar cells treated with TNF-alpha exhibited significantly higher MUC1 protein levels in detergent lysates compared with cells treated with vehicle alone. Increased MUC1 protein levels were correlated with significantly higher levels of MUC1 mRNA in TNF-alpha-treated cells compared with controls. However, TNF-alpha did not alter MUC1 transcript stability, implying increased de novo transcription induced by the cytokine. TNF-alpha increased MUC1 gene promoter activity in A549 cells transfected with a promoter-luciferase reporter plasmid. Both U0126, an inhibitor of MEK1/2, and dominant negative ERK1 prevented TNF-alpha-induced MUC1 promoter activation, and anti-TNFR1 antibody blocked TNF-alpha-stimulated ERK1/2 activation. MUC1 promoter activation by TNF-alpha also was blocked by mithramycin A, an inhibitor of Sp1, as well as either deletion or mutation of a putative Sp1 binding site in the MUC1 promoter located between nucleotides -99 and -90. TNF-alpha-stimulated binding of Sp1 to the MUC1 promoter in intact cells was demonstrated by chromatin immunoprecipitation assay. We conclude that TNF-alpha induces MUC1 gene transcription through a TNFR1 --> MEK1/2 --> ERK1 --> Sp1 pathway.


Subject(s)
Antigens, Neoplasm/genetics , Epithelial Cells/metabolism , Lung/cytology , Lung/metabolism , Mucins/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antigens, Neoplasm/metabolism , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung/drug effects , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Mucin-1 , Mucins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Sp1 Transcription Factor/metabolism , Time Factors
18.
Biochim Biophys Acta ; 1773(7): 1028-38, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17524503

ABSTRACT

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.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Proliferation , Mucins/metabolism , beta Catenin/metabolism , Amino Acid Sequence , Animals , Antigens, Neoplasm/genetics , CD8 Antigens/genetics , CD8 Antigens/metabolism , Cadherins/metabolism , Cells, Cultured , Cyclin D1/metabolism , Humans , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Molecular Sequence Data , Mucin-1 , Mucins/genetics , Protein Binding , Proto-Oncogene Proteins c-myc/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Transcription, Genetic
19.
Am J Physiol Lung Cell Mol Physiol ; 291(3): L407-16, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16632517

ABSTRACT

In this study, we investigated the regulation and mechanism of IL-8 expression by A549 human lung carcinoma cells treated with neutrophil elastase (NE). NE-treated cells exhibited significantly higher IL-8 protein levels in culture media compared with cells treated with vehicle alone. Blocking of gene transcription with actinomycin D suggested that NE stimulated IL-8 synthesis via increased mRNA expression, which was verified by real-time RT-PCR. NE activated the IL-8 promoter but did not alter the stability of its mRNA, confirming that the protease induced IL-8 synthesis through increased gene transcription. The results from the use of chemical inhibitors and mutant gene constructs against various signal transduction components seem to suggest the linear signaling pathway involving the activation of PKC-delta --> dual oxidase 1 --> reactive oxygen species --> TNF-alpha-converting enzyme --> EGF receptor --> p38 --> NF-kappaB for NE-activated IL-8 gene expression. A NF-kappaB potential binding site, located between nucleotides -82 and -69 of the IL-8 promoter, was identified as necessary for NE-induced IL-8 transcription. We conclude that NE increases IL-8 transcription through p38/NF-kappaB activation via EGFR transactivation.


Subject(s)
ErbB Receptors/metabolism , Interleukin-8/metabolism , Leukocyte Elastase/pharmacology , Lung/metabolism , NF-kappa B/metabolism , Transcriptional Activation , Binding Sites , Cell Line , Dose-Response Relationship, Drug , Epithelial Cells , ErbB Receptors/antagonists & inhibitors , Humans , Interleukin-8/genetics , Lung/cytology , Promoter Regions, Genetic , Protein Kinase C-delta/metabolism , RNA Stability , Signal Transduction , Transcription, Genetic/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
20.
J Immunol ; 176(7): 3890-4, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16547220

ABSTRACT

MUC1 (MUC1 in human and Muc1 in nonhumans) is a membrane-tethered mucin that interacts with Pseudomonas aeruginosa (PA) through flagellin. In this study, we compared PA pulmonary clearance and proinflammatory responses by Muc1(-/-) mice with Muc1(+/+) littermates following intranasal instillation of PA or flagellin. Compared with Muc1(+/+) mice, Muc1(-/-) mice showed increased PA clearance, greater airway recruitment of neutrophils, higher levels of TNF-alpha and KC in bronchoalveolar lavage fluid, higher levels of TNF-alpha in media of flagellin-stimulated alveolar macrophages, and higher levels of KC in media of tracheal epithelial cells. Knockdown of MUC1 enhanced flagellin-induced IL-8 production by primary human bronchial epithelial cells. Expression of MUC1 in HEK293T cells attenuated TLR5-dependent IL-8 release in response to flagellin, which was completely ablated when its cytoplasmic tail was deleted. We conclude that MUC1/Muc1 suppresses pulmonary innate immunity and speculate its anti-inflammatory activity may play an important modulatory role during microbial infection.


Subject(s)
Lung/immunology , Lung/microbiology , Mucin-1/genetics , Mucin-1/metabolism , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Animals , Cells, Cultured , Cytokines/biosynthesis , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Flagellin/pharmacology , Gene Deletion , Gene Expression Regulation/drug effects , Humans , Inflammation Mediators/metabolism , Mice , Mice, Knockout , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Pseudomonas Infections/genetics , RNA, Small Interfering/genetics , Toll-Like Receptor 5/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...