American Journal of Respiratory Cell and Molecular Biology

In this review, we summarize work over the past 15 years on mucin gene expression and regulation in the lung, as well as how mucin gene expression is altered in chronic lung diseases. This field owes a great debt to Carol Basbaum for her pioneering work in dissecting signaling pathways regulating mucin gene expression and for her tremendous energy in promoting the importance of understanding the basic pathogenic mechanisms that drive mucus overproduction in cystic fibrosis, chronic obstructive pulmonary disease, and asthma.

The luminal surface of the airway is coated by mucus, a protective barrier against toxins and pathogens, which clears particles and infectious agents from the airways via mucociliary clearance, an important component of the innate immune system of the lungs (1). Mucus is composed of water, ions, proteins, lipids, and glycoproteins. Mucin glycoproteins are a major macromolecular constituent of normal mucus, comprising 40–50% by weight of dialyzed, lyophilized mucus from healthy individuals (2). In cystic fibrosis (CF), asthma, and chronic obstructive pulmonary disease (COPD), the biochemical constituents of mucus are more complex (3).

With chronic infection and/or inflammation, cells and cellular components are included in mucus, creating sputum that contains leukocytes, DNA, proteoglycans, and filamentous actin. Further complicating the sputum mixture are fragments of mucins and other glycoproteins that are digested by leukocyte and bacterial proteases. Historically, biochemical and histochemical analyses revealed that mucins are present at high levels in sputum of patients with CF (47), chronic bronchitis (8), and asthma (9), and are a major component of sputum. A few reports indicate that the relative concentration of mucins in sputum is decreased compared with normal mucus due to degradation of mucins or increased amounts of other proteins and proteoglycans (2, 10). Overall, studies to evaluate changes in mucins in lung disease are very limited because purifying mucin glycoproteins is technically difficult due to their viscoelastic properties and heterogeneous structures. Ascertaining primary changes in mucin levels in lung disease has been complicated by secondary effects of proteases and glycosidases in infected sputum. A significant breakthrough in the field was the cloning of mucin genes, which encode the protein backbone of mucins. These studies, starting in the late 1980s and early 1990s, permitted evaluation of mucin gene expression in disease states, unperturbed by proteolytic degradation or by the heterogeneity of sputum constituents.

MUC1 was the first mucin gene cloned and revealed significant information concerning mucin expression and molecular structure. MUC1 was cloned from breast cancer tissue (11) and from pancreatic tumor cells (12). MUC1 cDNA encodes a large domain of tandemly repeating amino acids (tandem repeat domain) rich in serine and threonine residues, which are the putative sites of O-linked glycosylation. Further determination of mucin gene sequences has established that the tandem repeat domains constitute the largest domain of the molecule and that there is no significant homology in tandem repeat sequence between different mucin genes or indeed between homologs of different species. However, there may be variability in size of a single mucin between different individuals because each allele may contain different numbers of repeats. In addition to the tandem repeat domain, MUC1 cDNA encodes a transmembrane domain. These studies demonstrated that there exists a previously unrecognized family of mucin glycoproteins that are tethered to the epithelial apical membrane. Other membrane-tethered glycoproteins that have serine/threonine-rich domains and O-glycosylation are known, but their lack of tandem repeat domains prevent them from being considered as mucin glycoproteins.

The first secreted mucin gene identified, MUC2, was originally cloned from intestinal epithelium (13). Secreted mucins likewise contain the signature feature of mucins, the large tandem repeat domain of amino acids rich in serine and threonine where O-linked glycans are anchored. However, instead of a transmembrane domain, they contain several characteristic modules at their amino and carboxyl termini, including domains with homology to the D domains of the Von Willebrand factor. The D domains are believed to be functionally important for oligomerization of mucin monomers likely affecting mucin biophysical properties. Detailed descriptions of the modular motifs of mucins can be found in two recent reviews (14, 15).

Although the first mucin genes were not cloned from respiratory tract tissues, Craig Gerard and coworkers (16) and Carol Basbaum and colleagues (17) were the first to demonstrate that epithelial mucins are also expressed in the lung by demonstrating that MUC2 mRNA is expressed in the respiratory tract. This was followed by detection of MUC1 mRNA in the respiratory tract (18). Further advancing the field of airway mucin biology were the isolation and characterization of mucin cDNA clones from nasal and airway tissues. These genes, MUC5AC (19) and MUC5B (20), are localized adjacent to each other on chromosome 11p15.5 and are recognized to be two major secreted mucin genes expressed in the respiratory tract.

To date, 18 different human mucin genes have been cloned; at least 12 are expressed as mRNA in the lower respiratory tract (reviewed in Ref. 21). In the healthy lung, MUC1 and MUC4 are expressed at the apical surface of the respiratory epithelium. MUC5AC and MUC2 tend to be expressed in goblet cells of the superficial airway epithelium, while MUC5B, MUC8, and MUC19 are expressed in mucosal cells of submucosal glands. MUC7, a small, secreted, non–cysteine-rich mucin and a subclass of mucin unto itself, is not well-expressed normally and when expressed is localized to serosal cells of submucosal glands. The other mucins shown to be expressed as mRNA in lung tissue include MUC11, MUC13, MUC15, and MUC20; the localization of these genes within different compartments or cells in the lung is not yet known. The availability of mucin gene cDNA sequences has revolutionized the field of mucin biology permitting the development of cDNA probes and antibodies specific to cognate sequences in mucin protein backbones. These tools have been critical in the evaluation of cell-specific expression of mucin genes and proteins, and have enabled the evaluation of mucin expression in healthy lung and in airway diseases.

A major insight/contribution, perhaps initially promulgated by Professor Lynne Reid subsequent to her extensive histopathologic studies of lung tissues (22) and subsequently championed by Carol Basbaum, is that inflammation activated mucin gene expression (23). This concept has had wide-reaching ramifications, as it focused attention on the importance of mucin gene regulation in airway diseases. Importantly, Basbaum and colleagues first brought attention to the role of bacterial products, which are prevalent in CF airways, in regulating mucin expression in epithelial cells. This work further promoted the emerging concept that mucin glycoproteins are an important part of the innate immune defense system in the lung.

A key development that propelled this field has been the cloning of promoter sequences for several respiratory tract mucin genes including MUC1 (24), MUC2 (25, 26), MUC5AC (27), MUC5B (28, 29), and MUC4 (30, 31). Promoter sequences used in reporter chimera constructs have been an extremely useful tool to evaluate gene regulation and intracellular signaling in epithelial cells in vitro. This approach has resulted in a large body of literature defining the regulation of mucin genes in epithelial cells in vitro (reviewed in Ref. 21). Seminal studies, which established the current paradigm of mucin gene regulatory pathways, were initially performed using the MUC2 promoter (32), and more recently confirmed for MUC5AC. MUC2 and MUC5AC have several common active cis-elements in their promoters, including NF-κB, Sp-1, and CREB, suggesting that they share common regulatory pathways.

Mucus obstruction of airways and chronic inflammation are both fundamental features of CF, COPD, and asthma. Disease exacerbations with increased mucus production are also associated with bacterial or viral infections. These clinical observations suggest a link between infection, inflammation, and mucus production. Over the past 15 years, investigators have provided evidence establishing that infectious and inflammatory mediators interact with host epithelial cells and activate intracellular signaling pathways resulting in selective regulation of specific mucin genes. The studies on human mucins have been performed in vitro in lung cancer cell lines or in cultured primary airway epithelial cells to evaluate the effect of single agents and to permit dissection of the activated signaling pathways. Primarily, MUC gene expression has been evaluated and very few reports have correlated the increased mRNA findings with increased specific MUC protein expression. As discussed above, mucin glycoproteins are notoriously difficult to isolate, identify, and quantitate. Even with the development of antibodies for specific mucin proteins, it has been difficult to correlate gene regulatory studies with demonstration of increased protein production. A major conundrum that persists today is that despite evidence that MUC2 gene expression is upregulated in CF airways (33), it has been difficult to demonstrate the presence of MUC2 glycoprotein in normal mucus (34) or CF sputum (35). This contrasts with several reports that MUC5AC and MUC5B are major mucin glycoproteins in sputum from patients with asthma (36, 37), CF (35), and COPD (8). Bacteria relevant to different chronic and acute airway diseases have been evaluated for their effect on mucin regulation. Pseudomonas aeruginosa LPS (33), P. aeruginosa flagellin (38), Staphylococcus aureus lipoteichoic acid (39), Bordetella pertussis (40), and Haemophilus influenza (41) all transcriptionally upregulate MUC2 gene expression. P. aeruginosa LPS (27), H. influenza (42), and Mycobacterium pneumoniae (43) have also been reported to upregulate MUC5AC expression. In addition to bacteria, viral dsRNA also upregulates MUC2 expression (44).

Several cytokines and chemokines relevant to asthma, CF, and COPD also regulate mucin gene expression. TNF-α (4548), IL-1β (47, 48), and IL-9 (49, 50) upregulate both MUC2 and MUC5AC mRNA expression, whereas IL-6 and IL-17 increase expression of both MUC5AC and MUC5B (51). Interestingly, this is one of the few reports of mediators regulating MUC5B expression, which is particularly surprising considering the large amount of MUC5B mucin protein detected in mucus and sputum.

Other major airway inflammatory mediators also increase MUC5AC expression in vitro, including prostaglandins (46); proteases such as neutrophil elastase (52, 53), human airway trypsin (54), tissue kallikrein (55), metalloproteinases (56); and reactive oxygen species (55, 57, 58). Toxic agents that induce airway inflammation and oxidant stress, including tobacco smoke and pollutants (46, 5961), also increase MUC5AC expression.

The diverse array of infectious and inflammatory factors described above mediate mucin gene transcription by activating cell surface receptors and signaling via a limited repertoire of intracellular pathways. The initial step in the process requires the activation of cell surface receptors specific for each stimulus—for example, Toll-like receptor 2 for H. influenzae (41), (42), platelet activating factor receptor for S. aureus lipoteichoic acid (39), asialoGM1 for P. aeruginosa flagellin (62), and P2Y2 receptor for dsRNA (44). Activation of the retinoic acid receptor α (47) and the epidermal growth factor receptor (53, 6365) have been implicated in mucin gene regulation following several different stimuli. The cell surface receptors signal via downstream pathways, usually resulting in direct or indirect activation of the mitogen-activating protein kinase system. The ultimate step leading to MUC2 or MUC5AC upregulation is the activation of several transcription factors including NF-κB, AP-1, Sp-1, or CREB (reviewed in Ref. 21).

Although most published reports on MUC gene regulation have examined transcriptional mechanisms of regulation, there is a growing body of literature providing evidence that post-transcriptional mechanisms of regulation are important in mediating MUC mRNA abundance. TNF-α (46), neutrophil elastase (52, 66) and IL-8 (67) increase MUC5AC mRNA stability. MUC2 mRNA is also increased by post-transcriptional mechanisms after epithelial exposure to 12-O-tetradecanoylphorbol-13-acetate or forskolin (68). MUC4 is post-translationally regulated by TGF-β in rat mammary epithelial cells (69) and post-transcriptionally regulated by neutrophil elastase in primary normal human bronchial epithelial cells (70). Mechanisms whereby mucin mRNA is stabilized are not yet known; future studies are required to determine the cis stability domains in the mRNA sequence, intracellular signals required for RNA-binding protein–mRNA interactions, and the identity of the RNA binding proteins required for mRNA stabilization.

Few studies on the downregulation of mucin gene expression have been reported, although clearly repression of mucin production in chronic airway diseases by pharmacologic agents or anti-inflammatory mediators would be clinically useful. The glucocorticoid, dexamethasone (Dex), decreases the abundance of MUC5AC mRNA in airway epithelial cell lines (7173), primary differentiated NHBE cells (73), and rat primary differentiated and undifferentiated airway epithelial cells (72). Recent data have demonstrated that the Dex-induced repression of MUC5AC mRNA abundance in airway epithelial cell lines is regulated at the transcriptional, not the post-transcriptional level, and that the ligand-activated glucocorticoid receptor binds to two GRE cis-sites in the MUC5AC promoter (73).

However, it is not yet established whether repression of mucin genes results in concomitant decrease of mucin production in vivo or in vitro. An inhaled glucorticoid reduced CA19–9, a mucin marker in the bronchoalveolar lavage of patients with CF (74). Dex represses mucin synthesis in airway epithelial cell lines (71), but repression of Muc5ac mRNA does not result in decreased production of Muc5ac mucin in differentiated rat airway epithelial cells (72). This has not yet been evaluated in primary differentiated NHBE cells. Identifying mechanisms that effect downregulation of mucin gene expression and mucin production will be important to better understand processes whereby mucus overproduction can be reversed and mucus obstruction averted or overcome.

Mouse models have been extremely useful to test hypotheses of asthma or COPD pathogenesis. Although mouse models are also used to assess mechanisms of mucin gene regulation, the resulting phenotype is far more complex because the stimuli tested typically induce goblet cell metaplasia, thereby resulting in increased MUC gene and protein expression (reviewed in Ref. 20). This is an important point, because the regulatory factors required for goblet cell metaplasia may be different than those required to upregulate MUC expression.

There are several features of murine airways that are different from human airways, which must be considered when interpreting in vivo data on mucin gene regulation. The major secretory cell in murine airways is the Clara cell (75); there are very few resident goblet cells in normal mice. Mice phenotypes vary dramatically by their background strain (76), therefore characteristics ascribed to transgenes or loss of gene function must be evaluated in light of their background. Finally, it has been very challenging to establish murine models of CF lung disease using CFTR-null or CFTR-mutant mice, as the mice do not develop airway disease spontaneously (77). The failure of “CF” mice to develop chronic bronchitis and increased mucus production probably is due to the presence of alternative ion channels in murine airways that compensate for loss of CFTR function (78). Despite these caveats, the use of murine models to study airway diseases gained tremendous momentum with the recognition that Th2 cytokine exposure or allergen sensitization recapitulated many features in mouse airways seen in asthmatic airways, including goblet cell metaplasia. Both cDNA probes and MUC-specific antibodies are available for murine Muc5ac, and many reports have established that Muc5ac expression, not normally present in mouse airways, correlates directly with Alcian blue–periodic acid Schiff staining, the histochemical marker for goblet cells (79, 80).

To date, many mediators that regulate human MUC5AC mRNA expression in vitro may also increase airway goblet cell number and Muc5ac expression in vivo. The observed increase in Muc5ac mRNA or protein expression likely reflects the increased number of goblet cells in mouse airways. Mediators that induce goblet cell metaplasia include cigarette smoke (81, 82), acrolein (83), neutrophil elastase (84), IL-1β (85), IL-6/ IL-17 (86), IL-9 (87), TNF-α (88), LPS (89), M. pneumoniae infection (43), and UTP (90). To confirm that these mediators regulate murine Muc5ac mRNA expression, mRNA steady-state equilibrium and promoter-reporter functional experiments need to be done in vitro with mouse airway epithelial cells. An exception to these reports is the contrasting effect of the Th2 cytokines, IL-13 and IL-4, on mouse lung in vivo versus exposure to human airway epithelial cells in vitro. IL-13 and IL-4 do not increase human MUC5AC mRNA expression (86), although IL-13 (91), and IL-4 (92) induce goblet cell metaplasia in vivo, thereby resulting in an increase in Muc5ac expression. This exception illustrates the principle that signaling pathways that regulate goblet cell metaplasia may be different than the signaling pathways required for MUC gene regulation, and may vary between mice and humans.

The concept that normal mucus has important innate immune properties is becoming an accepted paradigm. However, there is still limited information on the functions attributable directly to the mucin glycoproteins, particularly the secreted mucins. The tremendous progress in understanding transcriptional mechanisms of mucin gene regulation still needs to be extended to the areas of in vivo regulation of Muc expression and of post-transcriptional mechanisms of MUC/Muc gene regulation. Finally, the promise of mucin research is the opportunity to discover new therapies to reduce mucin production and goblet cell hyperplasia. This hope has not yet been actualized and is the driving objective of mucin research for many investigators following the vision of Carol Basbaum.

1. Corfield AP, Shukla AK. Mucins: vital components of the mucosal defensive barrier. Genomic/Proteomic Technology 2003;3:20–22.
2. Rose MC, Brown CF, Jacoby JZ III, Lynn WS, Kaufman B. Biochemical properties of trachobronchial mucins from cystic fibrosis and non-cystic fibrosis individuals. Pediatr Res 1987;22:545–551.
3. Rubin BK, Ramirez O, Zayas JG, Finegan B, King M. Collection and analysis of respiratory mucus from subjects without lung disease. Am Rev Respir Dis 1990;141:1040–1043.
4. Zuelzer WW, Newton WA Jr. The pathogenesis of fibrocystic diseases of the pancreas: a study of 36 cases with special reference to the pulmonary lesions. Pediatrics 1949;4:53.
5. Boat TF, Cheng PW, Wood RE. Tracheobronchial mucus secretion in vivo and in vitro by epithelial tissues from cystic fibrosis and control subjects. Mod Probl Paediatr 1976;19:141–152.
6. Boat TF, Cheng PW, Iyer RN, Carlson DM, Polony I. Human respiratory tract secretions: mucous glycoproteins of nonpurulent tracheobronchialsecretions, and sputum of patients with bronchitis and cystic fibrosis. Arch Biochem Biophys 1976;177:95–104.
7. Lamblin G, Lafitte JJ, Lhermitte M, Degand P, Roussel P. Mucins from cystic fibrosis sputum. Mod Probl Paediatr 1976;19:153–164.
8. Caramori G, Di Gregorio C, Carlstedt I, Casolari P, Guzzinati I, Adcock IM, Barnes PJ, Ciaccia A, Cavallesco G, Chung KF, et al. Mucin expression in peripheral airways of patients with chronic obstructive pulmonary disease. Histopathology 2004;45:477–484.
9. Fahy JV, Steiger DJ, Liu J, Basbaum CB, Finkbeiner WE, Boushey HA. Markers of mucus secretion and DNA levels in induced sputum from asthmatic and from healthy subjects. Am Rev Respir Dis 1993;147:1132–1137.
10. Henke M, Renner A, Huber RM, Seeds MC, Rubin BK. MUC5AC and MUC5B mucins are decreased in cystic fibrosis airway secretions. Am J Respir Cell Mol Biol 2004;31:86–91.
11. Gendler SJ, Lancaster CA, Taylor-Papadimitriou J, Duhig T, Peat N, Burchell J, Pemberton L, Lalani EN, Wilson D. Molecular cloning and expression of human tumor-associated polymorphic epithelial mucin. J Biol Chem 1990;265:15286–15293.
12. Lan MS, Batra SK, Qi WN, Metzgar RS, Hollingsworth MA. Cloning and sequencing of a human pancreatic tumor mucin cDNA. J Biol Chem 1990;265:15294–15299.
13. Gum JR Jr, Byrd JC, Hicks JW, Toribara NW, Lamport DTA, Kim YS. Molecular cloning of human intestinal mucin cDNAs. J Biol Chem 1989;264:6480–6487.
14. Dekker J, Rossen JWA, Iler HAB, Einerhand AWC. The MUC family: an obituary. Trends Biochem Sci 2002;27:126–131.
15. Hollingsworth MA, Swanson BJ. Mucins in cancer: protection and control of the cell surface. Nat Rev Cancer 2004;4:45–60.
16. Gerard C, Eddy RL Jr, Shows TB. The core polypeptide of cystic fibrosis tracheal mucin contains a tandem repeat structure: evidence for a common mucin in airway and gastrointestinal tissue. J Clin Invest 1990;86:1921–1927.
17. Jany BH, Gallup MW, Yan PS, Gum JR Jr, Kim YS, Basbaum CB. Human bronchus and intestine express the same mucin gene. J Clin Invest 1991;87:77–82.
18. Hollingsworth MA, Batra SK, Qi WN, Yankaskas JR. MUC1 mucin mRNA expression in cultured human nasal and bronchial eptihelial cells. Am J Respir Cell Mol Biol 1992;6:516–520.
19. Meerzaman D, Charles P, Daskal E, Polymeropoulos MH, Martin BM, Rose MC. Cloning and analysis of cDNA encoding a major airway glycoprotein, human tracheobronchial mucin (MUC5). J Biol Chem 1994;269:12932–12939.
20. Desseyn JL, Guyonnet-Duperat V, Porchet N, Aubert JP, Laine A. Human mucin gene MUC5B, the 10.7 kb large central exon encodes various alternate subdomains resulting in a super-repeat. J Biol Chem 1997;272:3168–3178.
21. Rose MC, Voynow JA. Respiratory tract mucin genes and mucin glycoproteins in health and disease. Physiol Rev 2006;86:245–278.
22. Reid LM. The pathology of obstructive and inflammatory airway diseases. Eur J Respir Dis 1986;147:26–37.
23. Basbaum C, Gallup M, Gum J Jr, Kim Y, Jany B. Modification of mucin gene expression in the airways of rats exposed to sulfur dioxide. Biorheology 1990;27:485–489.
24. Lancaster CA, Peat N, Duhig T, Wilson D, Taylor-Papadimitriou J, Gendler SJ. Structure and expression of the human polymorphic epithelial mucin gene: an expressed VNTR unit. Biochem Biophys Res Commun 1990;173:1019–1029.
25. Velcich A, Palumbo L, Selleri L, Evans G, Augenlicht L. Organization and regulatory aspects of the human intestinal mucin gene (MUC2) locus. J Biol Chem 1997;272:7968–7976.
26. Gum JR, Hicks JW, Kim YS. Identification and characterization of the MUC2 (human intestinal mucin) gene 5′-flanking region: promoter activity in cultured cells. Biochem J 1997;325:259–267.
27. Li D, Gallup M, Fan N, Szymkowski DE, Basbaum CB. Cloning of the amino-terminal and 5′-flanking region of the human MUC5AC mucin gene and transcriptional up-regulation by bacterial exoproducts. J Biol Chem 1998;273:6812–6820.
28. Van Seuningen I, Perrais M, Pigny P, Porchet N, Aubert JP. Sequence of the 5′-flanking region and promoter activity of the human mucin gene MUC5B in different phenotypes of colon cancer cells. Biochem J 2000;348:675–686.
29. Chen Y, Zhao YH, Di Y-P, Wu R. Characterization of human mucin 5B gene expression in airway epithelium and the genomic clone of the amino-terminal and 5′-flanking region. Am J Respir Cell Mol Biol 2001;25:542–553.
30. Price-Schiavi SA, Perez A, Barco R, Carraway KL. Cloning and characterization of the 5′ flanking region of the sialomucin complex/rat Muc4 gene: promoter activity in cultured cells. Biochem J 2000;349:641–649.
31. Perrais M, Pigny P, Ducourouble M-P, Petitprez D, Porchet N, Aubert J-P, Van Seuningen I. Characterization of human mucin gene MUC4 promoter. Importance of growth factors and proinflammatory cytokines for its regulation in pancreatic cancer cells. J Biol Chem 2001;276:30923–30933.
32. Li JD, Feng W, Gallup M, Kim JH, Gum J, Kim Y, Basbaum C. Activation of NF-κB via a Src-dependent Ras-MAPK-pp90rsk pathway is required for Pseudomonas aeruginosa-induced mucin overproduction in epithelial cells. Proc Natl Acad Sci USA 1998;95:5718–5723.
33. Li J-D, Dohrman AF, Gallup M, Miyata S, Gum JR, Kim YS, Nadel JA, Prince A, Basbaum CB. Transcriptional activation of mucin by Pseudomonas aeruginosa lipopolysaccharide in the pathogenesis of cystic fibrosis lung disease. Proc Natl Acad Sci USA 1997;94:967–972.
34. Hovenberg HW, Davies JR, Herrmann A, Linden C-J, Carlstedt I. MUC5AC, but not MUC2, is a prominent mucin in respiratory secretions. Glycoconj J 1996;13:839–847.
35. Davies JR, Svitacheva N, Lannefors L, Kornfalt R, Carlstedt I. Identification of MUC5B, MUC5AC and small amounts of MUC2 mucins in cystic fibrosis airway secretions. Biochem J 1999;344:321–330.
36. Rose MC, Kaufman B, Martin BM. Proteolytic fragmentation and peptide mapping of human carboxyamidomethylated tracheobronchial mucin. J Biol Chem 1989;264:8193–8199.
37. Thornton D, Howard M, Khan N, Sheehan JK. Identification of two glycoforms of the MUC5B mucin in human respiratory mucus. J Biol Chem 1997;272:9561–9566.
38. McNamara N, Khong A, McKemy D, Caterina M, Boyer J, Julius D, Basbaum C. ATP transduces signals from ASGM1, a glycolipid that functions as a bacterial receptor. Proc Natl Acad Sci USA 2001;98:9086–9091.
39. Lemjabbar H, Basbaum C. Platelet-activating factor receptor and ADAM10 mediate responses to Staphylococcus aureus in epithelial cells. Nat Med 2002;8:41–46.
40. Belcher CE, Drenkow J, Kehoe B, Gingeras TR, McNamara N, Lemjabbar H, Basbaum C, Relman DA. The transcriptional responses of respiratory epithelial cells to Bordetella pertussis reveal host defensive and pathogen counter-defensive strategies. Proc Natl Acad Sci USA 2000;97:13847–13852.
41. Jono H, Shuto T, Xu H, Kai H, Lim DJ, Gum JR Jr, Kim YS, Yamaoka S, Feng X-H, Li J-D. Transforming growth factor-beta-Smad signaling pathway cooperates with NF-kB to mediate nontypeable Haemophilus influenzae-induced MUC2 mucin transcription. J Biol Chem 2002;277:45547–45557.
42. Wang B, Lim DJ, Han J, Kim YS, Basbaum CB, Li J-D. Novel cytoplasmic proteins of nontypeable Haemophilus influenzae up-regulate human MUC5AC mucin transcription via a positive p38 mitogen-activated protein kinase pathway and a negative phosphoinositide 3-kinase-Akt pathway. J Biol Chem 2002;277:949–957.
43. Chu HW, Jeyaseelan S, Rino JG, Voelker DR, Wexler RB, Campbell K, Harbeck RJ, Martin RJ. TLR2 signaling is critical for Mycoplasma pneumoniae-induced airway mucin expression. J Immunol 2005;174:5713–5719.
44. Londhe V, McNamara N, Lemjabbar H, Basbaum C. Viral dsRNA activates mucin transcription in airway epithelial cells. FEBS Lett 2003;553:33–38.
45. Levine SJ, Larivée P, Logun C, Angus CW, Ognibene FP, Shelhamer JH. Tumor necrosis factor-α induces mucin hypersecretion and MUC-2 gene expression by human airway epithelial cells. Am J Respir Cell Mol Biol 1995;12:196–204.
46. Borchers MT, Carty MP, Leikauf GD. Regulation of human airway mucins by acrolein and inflammatory mediators. Am J Physiol 1999;276:L549–L555.
47. Koo J, Kim YD, Jetten AM, Belloni P, Nettesheim P. Overexpression of mucin genes induced by interleukin-1beta, tumor necrosis factor-alpha, lipopolysacharide, and neutrophil elastase is inhibited by a retinoic acid receptor alpha antagonist. Exp Lung Res 2002;28:315–332.
48. Song K, Lee W-J, Chung KC, Koo JS, Yang EJ, Choi JY, Yoon J-H. Interleukin-1beta and tumor necrosis factor-alpha induce MUC5AC overexpression through a mechanism involving ERK/p38 mitogen-activated protein kinases-MSK1-CREB activation in human airway epithelial cells. J Biol Chem 2003;278:23243–23250.
49. Longphre M, Li D, Gallup M, Drori E, Ordoñez CL, Redman T, Wenzel S, Bice DE, Fahy JV, Basbaum C. Allergen-induced IL-9 directly stimulates mucin transcription in respiratory epithelial cells. J Clin Invest 1999;104:1375–1382.
50. Louahed J, Toda M, Jen J, Hamid Q, Renauld J-C, Levitt RC, Nicolaides NC. Interleukin-9 upregulates mucus expression in the airways. Am J Respir Cell Mol Biol 2000;22:649–656.
51. Chen Y, Thai P, Zhao Y-H, Ho Y-S, DeSouza MM, Wu R. Stimulation of ariway mucin gene expression by interleukin (IL)-17 through IL-6 paracrine/autocrine loop. J Biol Chem 2003;278:17036–17043.
52. Voynow JA, Young LR, Wang Y, Horger T, Rose MC, Fischer BM. Neutrophil elastase increases MUC5AC mRNA and protein expression in respiratory epithelial cells. Am J Physiol 1999;276:L835–L843.
53. Kohri K, Ueki IF, Nadel JA. Neutrophil elastase induces mucin production by ligand-dependent epidermal growth factor receptor activation. Am J Physiol Lung Cell Mol Physiol 2002;283:L531–L540.
54. Chokki M, Yamamura S, Eguchi H, Masegi T, Horiuchi H, Tanabe H, Kamimura T, Yasuoka S. Human airway trypsin-like protease increases mucin gene expression in airway epithelial cells. Am J Respir Cell Mol Biol 2004;30:470–478.
55. Casalino-Matsuda S, Monzon ME, Conner GE, Salathe M, Forteza RM. Role of hyaluronan and reactive oxygen species in tissue kallikrein-mediated epidermal growth factor receptor activation in human airways. J Biol Chem 2004;279:21606–21616.
56. Deshmukh HS, Case LM, Wesselkamper SC, Borchers MT, Martin LD, Shertzer HG, Nadel JA, Leikauf GD. Metalloproteinases mediate mucin 5AC expression by epidermal growth factor receptor activation. Am J Respir Crit Care Med 2005;171:305–314.
57. Takeyama K, Dabbagh K, Jeong Shim J, Dao-Pick T, Ueki IF, Nadel JA. Oxidative stress causes mucin synthesis via transactivation of epidermal growth factor receptor: role of neutrophils. J Immunol 2000;164:1546–1552.
58. Fischer BM, Voynow JA. Neutrophil elastase induces MUC5AC gene expression in airway epithelium via a pathway involving reactive oxygen species. Am J Respir Cell Mol Biol 2002;26:447–452.
59. Longphre M, Li D, Li J, Matovinovic E, Gallup M, Samet JM, Basbaum CB. Lung mucin production is stimulated by the air pollutant residual oil fly ash. Toxicol Appl Pharmacol 2000;162:86–92.
60. Gensch E, Gallup M, Sucher A, Li D, Gebremichael A, Lemjabbar H, Mengistab A, Dasari V, Hotchkiss J, Harkema J, et al. Tobacco smoke control of mucin production in lung cells requires oxygen radicals AP-1 and JNK. J Biol Chem 2004;279:39085–39093.
61. Shao MX, Nakanaga T, Nadel JA. Cigarette smoke induces MUC5AC mucin overproduction via tumor necrosis factor-alpha-converting enzyme in human airway epithelial cells (NCI-H292). Am J Physiol Lung Cell Mol Physiol 2004;287:L420–L427.
62. McNamara N, Khong A, McKemy D, Caterina M, Boyer J, Julius D, Basbaum C. ATP transduces signals from ASGM1, a glycolipid that functions as a bacterial receptor. Proc Natl Acad Sci USA 2001;98:9086–9091.
63. Perrais M, Pgny P, Copin M-C, Aubert J-P, Van Seuningen I. Induction of MUC2 and MUC5AC mucins by factors of the epidermal growth factor (EGF) family is mediated by EGF receptor/Ras/Raf/Extracellular signal-regulated kinase cascade and Sp1. J Biol Chem 2002;277:32258–32267.
64. Takeyama K, Dabbagh K, Jeong Shim J, Dao-Pick T, Ueki IF, Nadel JA. Oxidative stress causes mucin synthesis via transactivation of epidermal growth factor receptor: role of neutrophils. J Immunol 2000;164:1546–1552.
65. Takeyama K, Dabbagh K, Lee H-M, Agustí C, Lausier JA, Ueki IF, Grattan KM, Nadel JA. Epidermal growth factor system regulates mucin production in airways. Proc Natl Acad Sci USA 1999;96:3081–3086.
66. Voynow JA, Kenan DJ, Fischer BM. Neutrophil elastase increases A549 cell protein binding to a cis stability domain within the 3′untranslated region of MUC5AC mucin mRNA [abstract]. Am J Respir Crit Care Med 2002;165:A468.
67. Bautista M, Huang L, Letwin N, Rose MC. IL-8 receptors in airway cells and IL-8 regulation of mucin genes in vitro [abstract]. Am J Respir Crit Care Med 2001;163:A995.
68. Velcich A, Augenlicht LH. Regulated expression of an intestinal mucin gene in HT29 colonic carcinoma cells. J Biol Chem 1993;268:13956–13961.
69. Price-Schiavi SA, Carothers Carraway CA, Fregien N, Carraway KL. Post-transcriptional regulation of milk membrane protein, the sialomucin complex (ascites sialoglycoprotein (ASGP)-1/ASGP-2, rat Muc4), by transforming growth factor β. J Biol Chem 1998;273:35228– 35237.
70. Fischer BM, Cuellar JG, Diehl ML, deFreytas AM, Zhang J, Carraway KL, Voynow JA. Neutrophil elastase increases MUC4 expression in normal human bronchial epithelial cells. Am J Physiol Lung Cell Mol Physiol 2003;284:L671–L679.
71. Kai H, Yoshitake K, Hisatsune A, Kido T, Isohama Y, Takahama K, Miayata T. Dexamethasone suppresses mucus production and MUC2 and MUC5AC gene expression by NCI-H292 cells. Am J Physiol 1996;271:L484–L488.
72. Lu W, Lillehoj E, Kim KC. Effects of Dexamethasone on Muc5ac mucin production by primary airway goblet cells. Am J Physiol Lung Cell Mol Physiol 2005;288:L52–L60.
73. Chen YA, Nickola TJ, DiFronzo N, Colberg-Poley AM, Rose MC. Dexmethasone-mediated repression of MUC5AC mucin gene expression in human lung epithelial cells. Am J Respir Cell Mol Biol 2006;34:1–10.
74. Wojtczak HA, Kerby GS, Wagener JS, Copenhaver SC, Gotlin RW, Riches DWH, Accurso FJ. Beclomethasone diproprionate reduced airway inflammation without adrenal suppression in young children with cystic fibrosis: a pilot study. Pediatr Pulmonol 2001;32:293–302.
75. Pack RJ, Al-Ugaily LH, Morris G. The cells of the tracheobronchial epithelium of the mouse: a quantitative light and electron microscope study. J Anat 1981;132:71–84.
76. Whitehead GS, Walker JK, Berman KG, Foster WM, Schwartz DA. Allergen-induced airway disease is mouse strain dependent. Am J Physiol Lung Cell Mol Physiol 2003;285:L32–L42.
77. Frizzell RA, Pilewski JM. Finally, mice with CF lung disease. Nat Med 2004;10:452–454.
78. Grubb BR, Paradiso AM, Boucher RC. Anomalies in ion transport in CF mouse tracheal epithelium. Am J Physiol 1994;267:C293–C300.
79. Zuhdi Alimam M, Piazza FM, Selby DM, Letwin N, Huang L, Rose MC. Muc-5/5ac mucin messenger RNA and protein expression is a marker of goblet cell metaplasia in murine airways. Am J Respir Cell Mol Biol 2000;22:253–260.
80. Shahzeidi S, Aujla P, Nickola T, Chen Y, Alimam MZ, Rose MC. Temporal analysis of goblet cells and mucin gene expression in murine models of allergic asthma. Exp Lung Res 2003;29:549–565.
81. Miller L, Foster WM, Dambach DM, Doebler D, McKinnon M, Killar L, Longphre M. A murine model of cigarette smoke-induced pulmonary inflammation using intranasally administered smoke-conditioned medium. Exp Lung Res 2002;28:435–455.
82. Bartalesi B, Cavarra E, Fineschi S, Lucattelli M, Lunghi B, Martorana PA, Lungarella G. Different lung responses to cigarette smoke in two strains of mice sensitive to oxidants. Eur Respir J 2005;25:15–22.
83. Borchers MT, Wesselkamper S, Wert SE, Shapiro SD, Leikauf GD. Monocyte inflammation augments acrolein-induced Muc5ac expression in mouse lung. Am J Physiol 1999;277:L489–L497.
84. Voynow JA, Fischer BM, Malarkey DE, Burch LH, Wong T, Longphre M, Ho SB, Foster WM. Neutrophil elastase induces mucus cell metaplasia in mouse lung. Am J Physiol Lung Cell Mol Physiol 2004;287:L1293–L1302.
85. Lappalainen U, Whitsett JA, Wert SE, Tichelaar JW, Bry K. Interleukin-1beta causes pulmonary inflammation, emphysema, and airway remodeling in the adult murine lung. Am J Respir Cell Mol Biol 2005;32:311–318.
86. Chen Y, Thai P, Zhao Y-H, Ho Y-S, DeSouza MM, Wu R. Stimulation of airway mucin gene expression by interleukin (IL)-17 through IL-6 paracrine/autocrine loop. J Biol Chem 2003;278:17036–17043.
87. Reader JR, Hyde DM, Schelegle ES, Aldrich MC, Stoddard AM, McLane MP, Levitt RC, Tepper JS. Interleukin-9 induces mucous cell metaplasia independent of inflammation. Am J Respir Cell Mol Biol 2003;28:664–672.
88. Busse PJ, Zhang TF, Srivastava K, Lin BP, Schofield B, Sealfon SC, Li XM. Chronic exposure to TNF-alpha increases airway mucus gene expression in vivo. J Allergy Clin Immunol 2005;116:1256–1263.
89. Yanagihara K, Seki M, Cheng P-W. Lipopolysaccharide induces mucus cell metaplasia in mouse lung. Am J Respir Cell Mol Biol 2001;24:66–73.
90. Chen Y, Zhao YH, Wu R. Differential regulation of airway mucin gene expression and mucin secretion by extracellular nucleotide triphosphates. Am J Respir Cell Mol Biol 2001;25:409–417.
91. Wills-Karp M, Luyimbazi J, Xu X, Schofield B, Neben TY, Karp CL, Donaldson DD. Interleukin-13: central mediator of allergic asthma. Science 1998;282:2258–2263.
92. Temann U, Prasad B, Gallup M, Basbaum C, Ho S, Flavell R, Rankin J. A novel role for murine IL-4 in vivo: induction of MUC5AC gene expression and mucin hypersecretion. Am J Respir Cell Mol Biol 1997;16:471–478.
Correspondence and requests for reprints should be addressed to Judith A. Voynow, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710. E-mail:

Related

No related items
American Journal of Respiratory Cell and Molecular Biology
34
6

Click to see any corrections or updates and to confirm this is the authentic version of record