Smoking-associated increase in mucins 1 and 4 in human airways

Heta Merikallio, Riitta Kaarteenaho, Sara Lindén, Médea Padra, Reza Karimi, Chuan-Xing Li, Elisa Lappi-Blanco, Åsa M Wheelock, Magnus C Sköld, Heta Merikallio, Riitta Kaarteenaho, Sara Lindén, Médea Padra, Reza Karimi, Chuan-Xing Li, Elisa Lappi-Blanco, Åsa M Wheelock, Magnus C Sköld

Abstract

Rationale: Smoking-related chronic obstructive pulmonary disease (COPD) is associated with dysregulated production of mucus. Mucins (MUC) are important both for mucus secretion and epithelial defense. We have examined the distribution of MUC1 and MUC4 in the airway epithelial cells of never-smokers and smokers with and without COPD.

Methods: Mucosal biopsies and bronchial wash samples were obtained by bronchoscopy from age- and sex-matched COPD-patients (n = 38; GOLD I-II/A-B), healthy never-smokers (n = 40) and current smokers with normal lung function (n = 40) from the Karolinska COSMIC cohort (NCT02627872). Cell-specific expressions of MUC1, MUC4 and regulating factors, i.e., epithelial growth factor receptor (EGFR) 1 and 2, were analyzed by immunohistochemistry. Soluble MUC1 was measured by quantitative immunodetection on slot blot.

Results: The levels of cell-bound MUC1 expression in basal cells and in soluble MUC1 in bronchial wash were increased in smokers, regardless of airway obstruction. Patients with chronic bronchitis had higher MUC1 expression. The expression of MUC4 in cells with goblet cell phenotype was increased in smokers. The expression of EGFR2, but not that of EGFR1, was higher in never-smokers than in smokers.

Conclusions: Smoking history and the presence of chronic bronchitis, regardless of airway obstruction, affect both cellular and soluble MUC1 in human airways. Therefore, MUC1 may be a novel marker for smoking- associated airway disease.

Keywords: Bronchus; COPD; Chronic Bronchitis; Epithelium; Mucin; Smoking.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Representative images of the immunohistochemical stainings for cell-bound MUC1 in bronchial biopsy samples. Intensity of the expression was designated as negative, faint, moderate, strong or very strong, and the extent of the positive staining was estimated from 0 to 100% in each cell type present in the airways. Expression of MUC1 in cells suggesting basal cell phenotype in the large airways of a Never-smoker with normal lung functions (a), a Smoker with normal lung function (b), MUC1 expression in basal cells of an ex-smoker with COPD (c) and a Smoker with COPD (d)
Fig. 2
Fig. 2
Immunohistochemical scores for cell-bound MUC1 were calculated based on the intensity and extent of the staining. Scores for immunohistochemical expression of MUC1 in basal cells of large airways from smokers (including COPD) and Never-smokers and from ex-smokers with COPD. MUC1 scores in all subjects (a), and scores separately in males (b) and females (c). Results are shown as mean bars with standard error of mean
Fig. 3
Fig. 3
Representative images of the immunohistochemical stainings for MUC4 in bronchial biopsy samples. MUC4 was positive mainly in cells suggesting basal cell and ciliated cell phenotypes, in some cases also in cells with goblet cell phenotype. MUC4 expression in ciliated and basal cells in a Never-smoker (a), a positive expression for MUC4 in the main epithelial cell types in a Smoker (b), an ex-smoker with COPD (c) and a smoker with COPD (d)
Fig. 4
Fig. 4
Significant results of the scores on the immunohistochemical expression of MUC4 in goblet cells of large airways. MUC4 scores in the goblet cells of all subjects (a) and in males (b). Results are shown as median bars with standard error of median
Fig. 5
Fig. 5
Representative images of the immunohistochemical stainings for EGFR1. Expression of EGFR in cells suggesting basal cell phenotype in the large airways of a Never-smoker with normal lung functions (a), a Smoker with normal lung function (b), MUC1 expression in basal cells of an ex-smoker with COPD (c) and a Smoker with COPD (d). Expression scores of EGFR1 in large airways (e)
Fig. 6
Fig. 6
Representative images of the immunohistochemical stainings for EGFR 2. Expression of EGFR2 in cells suggesting basal cell and ciliated cell phenotype in the large airways of a Never-smoker with normal lung functions (a), a Smoker with normal lung function (b), MUC1 expression in basal cells of an ex-smoker with COPD (c) and a Smoker with COPD (d). EGFR2 scores of all subjects in basal cell phenotype (e) and in ciliated cell phenotype (f). Results are shown as median bars and standard error of median
Fig. 7
Fig. 7
Soluble MUC1 from basal cells or sub-epithelial glands were measured with quantitative immunoblotting. MUC1 concentration in bronchial wash samples. MUC1 concentration in the whole group (a) and in the females (b). 1 unit equals 0.4 μg/mL. Results are shown as mean and standard error of mean
Fig. 8
Fig. 8
Cell-bound and soluble MUC1 as well as MUC4 was studied in large airways of non-smokers and smokers. The non-smoker group includes both never-smokers and COPD ex-smokers whereas the smoker group includes smokers with normal lung function and smokers with COPD. a MUC1 scores in cells suggesting basal cell phenotype from immunohistochemical stainings; b MUC4 scores in cells suggesting goblet cell phenotype from immunohistochemical stainings and c. Amount of MUC1 in bronchial wash samples; 1 unit equals 0.4 μg/mL. MUC1 results are shown as mean bars and standard error of mean, MUC4 results are shown as median bars and standard error of median
Fig. 9
Fig. 9
Differences in MUC1 expression levels in patients with or without chronic bronchitis. a Scores of cell-bound MUC1 expression in large airways and b. Amount of MUC1 in bronchial wash samples from the patients; 1 unit equals 0.4 μg/mL. MUC1 results are shown as mean bars and standard error, MUC1 results are shown as median bars and standard error of median

References

    1. Zhou-Suckow Z, Duerr J, Hagner M, Agrawal R, Mall MA. Airway mucus, inflammation and remodeling: emerging links in the pathogenesis of chronic lung diseases. Cell Tissue Res. 2017;367(3):537–550. doi: 10.1007/s00441-016-2562-z.
    1. Kesimer M, Ford AA, Ceppe A, Radicioni G, Cao R, Davis CW, et al. Airway Mucin Concentration as a Marker of Chronic Bronchitis. N Engl J Med. 2017;377(10):911–922. doi: 10.1056/NEJMoa1701632.
    1. Tian PW, Wen FQ. Clinical significance of airway mucus hypersecretion in chronic obstructive pulmonary disease. J Transl Int Med. 2015;3(3):89–92. doi: 10.1515/jtim-2015-0013.
    1. Wedzicha JA. Airway Mucins in Chronic Obstructive Pulmonary Disease. N Engl J Med. 2017;377(10):986–987. doi: 10.1056/NEJMe1707210.
    1. Corfield AP. Mucins: a biologically relevant glycan barrier in mucosal protection. Biochim Biophys Acta. 2015;1850(1):236–252. doi: 10.1016/j.bbagen.2014.05.003.
    1. Rose MC, Voynow JA. Respiratory tract mucin genes and mucin glycoproteins in health and disease. Physiol Rev. 2006;86(1):245–278. doi: 10.1152/physrev.00010.2005.
    1. Ramsey KA, Rushton ZL, Ehre C. Mucin Agarose Gel Electrophoresis: Western Blotting for High-molecular-weight Glycoproteins. J Vis Exp. 2016;(112):54153.
    1. Rose MC, Nickola TJ, Voynow JA. Airway mucus obstruction: mucin glycoproteins, MUC gene regulation and goblet cell hyperplasia. Am J Respir Cell Mol Biol. 2001;25(5):533–537. doi: 10.1165/ajrcmb.25.5.f218.
    1. Bafna S, Kaur S, Batra SK. Membrane-bound mucins: the mechanistic basis for alterations in the growth and survival of cancer cells. Oncogene. 2010;29(20):2893–2904. doi: 10.1038/onc.2010.87.
    1. Hattrup CL, Gendler SJ. Structure and function of the cell surface (tethered) mucins. Annu Rev Physiol. 2008;70:431–457. doi: 10.1146/annurev.physiol.70.113006.100659.
    1. Nath S, Mukherjee P. MUC1: a multifaceted oncoprotein with a key role in cancer progression. Trends Mol Med. 2014;20(6):332–342. doi: 10.1016/j.molmed.2014.02.007.
    1. Engelmann K, Kinlough CL, Muller S, Razawi H, Baldus SE, Hughey RP, et al. Transmembrane and secreted MUC1 probes show trafficking-dependent changes in O-glycan core profiles. Glycobiology. 2005;15(11):1111–1124. doi: 10.1093/glycob/cwi099.
    1. Linden SK, Sheng YH, Every AL, Miles KM, Skoog EC, Florin TH, et al. MUC1 limits Helicobacter pylori infection both by steric hindrance and by acting as a releasable decoy. PLoS Pathog. 2009;5(10):e1000617. doi: 10.1371/journal.ppat.1000617.
    1. Lapperre TS, Sont JK, van Schadewijk A, Gosman MM, Postma DS, Bajema IM, et al. Smoking cessation and bronchial epithelial remodelling in COPD: a cross-sectional study. Respir Res. 2007;8:85. doi: 10.1186/1465-9921-8-85.
    1. O'Donnell RA, Richter A, Ward J, Angco G, Mehta A, Rousseau K, et al. Expression of ErbB receptors and mucins in the airways of long term current smokers. Thorax. 2004;59(12):1032–1040. doi: 10.1136/thx.2004.028043.
    1. Takeyama K, Jung B, Shim JJ, Burgel PR, Dao-Pick T, Ueki IF, et al. Activation of epidermal growth factor receptors is responsible for mucin synthesis induced by cigarette smoke. Am J Physiol Lung Cell Mol Physiol. 2001;280(1):L165–L172. doi: 10.1152/ajplung.2001.280.1.L165.
    1. Patton S, Gendler SJ, Spicer AP. The epithelial mucin, MUC1, of milk, mammary gland and other tissues. Biochim Biophys Acta. 1995;1241(3):407–423. doi: 10.1016/0304-4157(95)00014-3.
    1. Lappi-Blanco E, Makinen JM, Lehtonen S, Karvonen H, Sormunen R, Laitakari K, et al. Mucin-1 correlates with survival, smoking status, and growth patterns in lung adenocarcinoma. Tumour Biol. 2016;37(10):13811–13820. doi: 10.1007/s13277-016-5269-6.
    1. Albrecht H, Carraway KL., 3rd MUC1 and MUC4: switching the emphasis from large to small. Cancer Biother Radiopharm. 2011;26(3):261–271. doi: 10.1089/cbr.2011.1017.
    1. Chaturvedi P, Singh AP, Chakraborty S, Chauhan SC, Bafna S, Meza JL, et al. MUC4 mucin interacts with and stabilizes the HER2 oncoprotein in human pancreatic cancer cells. Cancer Res. 2008;68(7):2065–2070. doi: 10.1158/0008-5472.CAN-07-6041.
    1. Balgoma D, Yang M, Sjodin M, Snowden S, Karimi R, Levanen B, et al. Linoleic acid-derived lipid mediators increase in a female-dominated subphenotype of COPD. Eur Respir J. 2016;47(6):1645–1656. doi: 10.1183/13993003.01080-2015.
    1. Forsslund H, Yang M, Mikko M, Karimi R, Nyren S, Engvall B, et al. Gender differences in the T-cell profiles of the airways in COPD patients associated with clinical phenotypes. Int J Chron Obstruct Pulmon Dis. 2017;12:35–48. doi: 10.2147/COPD.S113625.
    1. Naz S, Kolmert J, Yang M, Reinke SN, Kamleh MA, Snowden S, et al. Metabolomics analysis identifies sex-associated metabotypes of oxidative stress and the autotaxin-lysoPA axis in COPD. Eur Respir J. 2017;49(6):1602322.
    1. Karimi R, Tornling G, Forsslund H, Mikko M, Wheelock A, Nyren S, et al. Lung density on high resolution computer tomography (HRCT) reflects degree of inflammation in smokers. Respir Res. 2014;15:23. doi: 10.1186/1465-9921-15-23.
    1. Kohler M, Sandberg A, Kjellqvist S, Thomas A, Karimi R, Nyren S, et al. Gender differences in the bronchoalveolar lavage cell proteome of patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2013;131(3):743–751. doi: 10.1016/j.jaci.2012.09.024.
    1. Yang M, Kohler M, Heyder T, Forsslund H, Garberg HK, Karimi R, et al. Long-term smoking alters abundance of over half of the proteome in bronchoalveolar lavage cell in smokers with normal spirometry, with effects on molecular pathways associated with COPD. Respir Res. 2018;19(1):40. doi: 10.1186/s12931-017-0695-6.
    1. Yang M, Kohler M, Heyder T, Forsslund H, Garberg HK, Karimi R, et al. Proteomic profiling of lung immune cells reveals dysregulation of phagocytotic pathways in female-dominated molecular COPD phenotype. Respir Res. 2018;19(1):39. doi: 10.1186/s12931-017-0699-2.
    1. Mikko M, Forsslund H, Cui L, Grunewald J, Wheelock AM, Wahlstrom J, et al. Increased intraepithelial (CD103+) CD8+ T cells in the airways of smokers with and without chronic obstructive pulmonary disease. Immunobiology. 2013;218(2):225–231. doi: 10.1016/j.imbio.2012.04.012.
    1. Naz S, Bhat M, Stahl S, Forsslund H, Skold CM, Wheelock AM, et al. Dysregulation of the Tryptophan Pathway Evidences Gender Differences in COPD. Metabolites. 2019;9(10):212.
    1. Li CX, Wheelock CE, Skold CM, Wheelock AM. Integration of multi-omics datasets enables molecular classification of COPD. Eur Respir J. 2018;51(5):1701930.
    1. Sandberg A, Skold CM, Grunewald J, Eklund A, Wheelock AM. Assessing recent smoking status by measuring exhaled carbon monoxide levels. PLoS One. 2011;6(12):e28864. doi: 10.1371/journal.pone.0028864.
    1. al S-HCHe. Definition and classification of chronic bronchitis for clinical and epidemiological purposes. A report to the Medical Research Council by their Committee on the Aetiology of Chronic Bronchitis. Lancet. 1965;1(7389):775–779.
    1. Forsslund H, Mikko M, Karimi R, Grunewald J, Wheelock AM, Wahlstrom J, et al. Distribution of T-cell subsets in BAL fluid of patients with mild to moderate COPD depends on current smoking status and not airway obstruction. Chest. 2014;145(4):711–722. doi: 10.1378/chest.13-0873.
    1. Lofdahl JM, Cederlund K, Nathell L, Eklund A, Skold CM. Bronchoalveolar lavage in COPD: fluid recovery correlates with the degree of emphysema. Eur Respir J. 2005;25(2):275–281. doi: 10.1183/09031936.05.00033504.
    1. Karimi R, Tornling G, Grunewald J, Eklund A, Skold CM. Cell recovery in bronchoalveolar lavage fluid in smokers is dependent on cumulative smoking history. PLoS One. 2012;7(3):e34232. doi: 10.1371/journal.pone.0034232.
    1. Lee HJ, Xu X, Choe G, Chung DH, Seo JW, Lee JH, et al. Protein overexpression and gene amplification of epidermal growth factor receptor in nonsmall cell lung carcinomas: Comparison of four commercially available antibodies by immunohistochemistry and fluorescence in situ hybridization study. Lung Cancer. 2010;68(3):375–382. doi: 10.1016/j.lungcan.2009.07.014.
    1. Landis JR, Koch GG. The measurement of observer agreement for categorical data. Biometrics. 1977;33(1):159–174. doi: 10.2307/2529310.
    1. Macao B, Johansson DG, Hansson GC, Hard T. Autoproteolysis coupled to protein folding in the SEA domain of the membrane-bound MUC1 mucin. Nat Struct Mol Biol. 2006;13(1):71–76. doi: 10.1038/nsmb1035.
    1. McAuley JL, Linden SK, Png CW, King RM, Pennington HL, Gendler SJ, et al. MUC1 cell surface mucin is a critical element of the mucosal barrier to infection. J Clin Invest. 2007;117(8):2313–2324. doi: 10.1172/JCI26705.
    1. Caramori G, Casolari P, Di Gregorio C, Saetta M, Baraldo S, Boschetto P, et al. MUC5AC expression is increased in bronchial submucosal glands of stable COPD patients. Histopathology. 2009;55(3):321–331. doi: 10.1111/j.1365-2559.2009.03377.x.
    1. Kovalenko S, Dorofieiev A. The content of mucin MUC-2, −3 and −4 antigens in the bronchial mucosa membrane of chronic obstructive pulmonary disease patients during acute exacerbation - initial report. Adv Respir Med. 2017;85(1):3–7. doi: 10.5603/ARM.2017.0002.
    1. Ishikawa N, Mazur W, Toljamo T, Vuopala K, Ronty M, Horimasu Y, et al. Ageing and long-term smoking affects KL-6 levels in the lung, induced sputum and plasma. BMC Pulm Med. 2011;11:22. doi: 10.1186/1471-2466-11-22.
    1. Zheng Z, Qi Y, Xu X, Jiang H, Li Z, Yang Q, et al. Sputum mucin 1 is increased during the acute phase of chronic obstructive pulmonary disease exacerbation. J Thorac Dis. 2017;9(7):1873–1882. doi: 10.21037/jtd.2017.06.63.
    1. Ishizaka A, Matsuda T, Albertine KH, Koh H, Tasaka S, Hasegawa N, et al. Elevation of KL-6, a lung epithelial cell marker, in plasma and epithelial lining fluid in acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol. 2004;286(6):L1088–L1094. doi: 10.1152/ajplung.00420.2002.
    1. Shigemura M, Nasuhara Y, Konno S, Shimizu C, Matsuno K, Yamguchi E, et al. Effects of molecular structural variants on serum Krebs von den Lungen-6 levels in sarcoidosis. J Transl Med. 2012;10:111. doi: 10.1186/1479-5876-10-111.
    1. Nathani N, Perkins GD, Tunnicliffe W, Murphy N, Manji M, Thickett DR. Kerbs von Lungren 6 antigen is a marker of alveolar inflammation but not of infection in patients with acute respiratory distress syndrome. Crit Care. 2008;12(1):R12. doi: 10.1186/cc6785.
    1. Tomita M, Ayabe T, Chosa E, Nose N, Nakamura K. Prognostic significance of preoperative serum Krebs von den Lungen-6 level in non-small cell lung cancer. Gen Thorac Cardiovasc Surg. 2016;64(11):657–661. doi: 10.1007/s11748-016-0706-4.
    1. Xu T, Li D, Wang H, Zheng T, Wang G, Xin Y. MUC1 downregulation inhibits non-small cell lung cancer progression in human cell lines. Exp Ther Med. 2017;14(5):4443–4447.
    1. Schilders KA, Eenjes E, van Riet S, Poot AA, Stamatialis D, Truckenmuller R, et al. Regeneration of the lung: Lung stem cells and the development of lung mimicking devices. Respir Res. 2016;17:44. doi: 10.1186/s12931-016-0358-z.
    1. Caramori G, Di Gregorio C, Carlstedt I, Casolari P, Guzzinati I, Adcock IM, et al. Mucin expression in peripheral airways of patients with chronic obstructive pulmonary disease. Histopathology. 2004;45(5):477–484. doi: 10.1111/j.1365-2559.2004.01952.x.
    1. Aryal S, Diaz-Guzman E, Mannino DM. COPD and gender differences: an update. Transl Res. 2013;162(4):208–218. doi: 10.1016/j.trsl.2013.04.003.
    1. Bitler BG, Goverdhan A, Schroeder JA. MUC1 regulates nuclear localization and function of the epidermal growth factor receptor. J Cell Sci. 2010;123(Pt 10):1716–1723. doi: 10.1242/jcs.062661.
    1. Anagnostis A, Neofytou E, Soulitzis N, Kampas D, Drositis I, Dermitzaki D, et al. Molecular profiling of EGFR family in chronic obstructive pulmonary disease: correlation with airway obstruction. Eur J Clin Investig. 2013;43(12):1299–1306. doi: 10.1111/eci.12178.
    1. de Boer WI, Hau CM, van Schadewijk A, Stolk J, van Krieken JHJM, Hiemstra PS. Expression of Epidermal Growth Factors and Their Receptors in the Bronchial Epithelium of Subjects With Chronic Obstructive Pulmonary Disease. Am J Clin Pathol. 2006;125(2):184–192. doi: 10.1309/W1AXKGT7UA37X257.
    1. Suzuki M, Wada H, Yoshino M, Tian L, Shigematsu H, Suzuki H, et al. Molecular characterization of chronic obstructive pulmonary disease-related non-small cell lung cancer through aberrant methylation and alterations of EGFR signaling. Ann Surg Oncol. 2010;17(3):878–888. doi: 10.1245/s10434-009-0739-3.
    1. Woodruff PG, Wolff M, Hohlfeld JM, Krug N, Dransfield MT, Sutherland ER, et al. Safety and efficacy of an inhaled epidermal growth factor receptor inhibitor (BIBW 2948 BS) in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010;181(5):438–445. doi: 10.1164/rccm.200909-1415OC.
    1. Karg A, Dinc ZA, Basok O, Ucvet A. MUC4 expression and its relation to ErbB2 expression, apoptosis, proliferation, differentiation, and tumor stage in non-small cell lung cancer (NSCLC) Pathol Res Pract. 2006;202(8):577–583. doi: 10.1016/j.prp.2006.04.002.
    1. Yang M, Kohler M, Heyder T, Forsslund H, Garberg HK, Karimi R, et al. Proteomic profiling of lung immune cells reveals dysregulation of phagocytotic pathways in female-dominated molecular COPD phenotype. Respiratory Research. 2018. In press.
    1. Zaretsky JZ, Barnea I, Aylon Y, Gorivodsky M, Wreschner DH, Keydar I. MUC1 gene overexpressed in breast cancer: structure and transcriptional activity of the MUC1 promoter and role of estrogen receptor alpha (ERalpha) in regulation of the MUC1 gene expression. Mol Cancer. 2006;5:57. doi: 10.1186/1476-4598-5-57.

Source: PubMed

3
Abonneren