CC16 levels correlate with cigarette smoke exposure in bronchial epithelial cells and with lung function decline in smokers

David Chi-Leung Lam, Hoi-Hin Kwok, Wai-Cho Yu, Fanny Wai-San Ko, Cheuk-Yin Tam, Arthur Chun-Wing Lau, Daniel Yee-Tak Fong, Mary Sau-Man Ip, David Chi-Leung Lam, Hoi-Hin Kwok, Wai-Cho Yu, Fanny Wai-San Ko, Cheuk-Yin Tam, Arthur Chun-Wing Lau, Daniel Yee-Tak Fong, Mary Sau-Man Ip

Abstract

Background: Club cell protein-16 (CC16) expression has been associated with smoking-related lung function decline. The study hypothesis was that CC16 expression in both serum and bronchial epithelium is associated with lung function decline in smokers, and exposure to cigarette smoke will lead to reduction in CC16 expression in bronchial epithelial cells.

Methods: In a cohort of community-based male Chinese subjects recruited for lung function test in 2000, we reassessed their lung function ten years later and measured serum levels of CC16. CC16 expression was further assayed in bronchial epithelium from endobronchial biopsies taken from an independent cohort of subjects undergoing autofluorescence bronchoscopy, and tested for correlation between CC16 immunostaining intensity and lung function. In an in-vitro model, bronchial epithelial cells were exposed to cigarette smoke extract (CSE), and the expression levels of CC16 were measured in bronchial epithelial cells before and after exposure to CSE.

Results: There was a significant association between FEV1 decline and serum CC16 levels in smokers. Expression of CC16 in bronchial epithelium showed significant correlation with FEV1/FVC. Bronchial epithelial cells showed significant decrease in CC16 expression after exposure to CSE, followed by a subsequent rise in CC16 expression upon removal of CSE.

Conclusions: Results of these clinical and laboratory investigations suggested that low serum CC16 was associated with smoking-related decline in lung function, demonstrated the first time in a Chinese cohort. The data also lend support to the putative role of CC16 in protection against smoking-related bronchial epithelial damage. (Abstract word count: 243) US CLINICAL TRIAL REGISTRY: NCT01185652 , first posted 20 August, 2010.

Keywords: Biomarkers; Forced expiratory volume; Lung function; Smoking; Spirometry.

Conflict of interest statement

Ethics approval and consent to participate

The study was approved by Institutional Review Board of the University of Hong Kong/Hong Kong Hospital Authority Hong Kong West Cluster (HKHA HKWC 08–428 and 09–120) and the research was carried out in accordance with the Declaration of Helsinki (2008). Informed written consents were obtained from all participating subjects.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interest.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
A study flow chart to show the recruitment of subjects
Fig. 2
Fig. 2
Immunostaining for CC16 expression in endobronchial biopsies showing (A) Negligible staining (scored as 0 with the same intensity in bronchial epithelium) in control and (B) Weak staining (scored as 1) and (C) Strong staining (scored as 2) of CC16 in the bronchial epithelium. Note: Immunostaining were scored with the bronchial epithelial cytoplasmic staining intensity, not the stromal or nuclear staining intensity
Fig. 3
Fig. 3
Effects of cigarette smoke extract (CSE) exposure on the expression levels of CC16 mRNA. Eight immortalized normal human bronchial epithelial cell lines were treated with CSE for 96 h after overnight serum starvation. CSE was then removed and washed with PBS for further culture for another 96 h in the absence of CSE. Data were shown as mean +/− standard deviation of eight cell lines and were analyzed by one-way ANOVA followed by Tukey’s multiple comparison tests. The p values for comparison of group means at 0 h vs 96 h, and 96 h vs 192 h, are also listed. * p < 0.001 vs PBS vehicle control
Fig. 4
Fig. 4
Cell viability monitoring with Trypan Blue and MTT assays for different cell lines during experiments

References

    1. Fletcher C, Peto R. The natural history of chronic airflow obstruction. Br Med J. 1977;1(6077):1645–1648. doi: 10.1136/bmj.1.6077.1645.
    1. Drummond MB, Hansel NN, Connett JE, Scanlon PD, Tashkin DP, Wise RA. Spirometric predictors of lung function decline and mortality in early chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2012;185(12):1301–1306. doi: 10.1164/rccm.201202-0223OC.
    1. Global Strategy for the Diagnosis . Management and prevention of COPD, global initiative for chronic obstructive lung disease (GOLD) 2016. 2016.
    1. Dickens JA, Lomas DA. CC-16 as a biomarker in chronic obstructive pulmonary disease. COPD. 2012;9(5):574–575. doi: 10.3109/15412555.2012.718926.
    1. Celli BR, Locantore N, Yates J, Tal-Singer R, Miller BE, Bakke P, Calverley P, Coxson H, Crim C, Edwards LD, et al. Inflammatory biomarkers improve clinical prediction of mortality in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2012;185(10):1065–1072. doi: 10.1164/rccm.201110-1792OC.
    1. Garcia-Rio F, Miravitlles M, Soriano JB, Munoz L, Duran-Tauleria E, Sanchez G, Sobradillo V, Ancochea J, Committee E-SS Systemic inflammation in chronic obstructive pulmonary disease: a population-based study. Respir Res. 2010;11:63. doi: 10.1186/1465-9921-11-63.
    1. Knabe L, Fort A, Chanez P, Bourdin A. Club cells and CC16: another “smoking gun”? (with potential bullets against COPD) Eur Respir J. 2015;45(6):1519–1520. doi: 10.1183/09031936.00010515.
    1. Guerra S, Halonen M, Vasquez MM, Spangenberg A, Stern DA, Morgan WJ, Wright AL, Lavi I, Tares L, Carsin AE, et al. Relation between circulating CC16 concentrations, lung function, and development of chronic obstructive pulmonary disease across the lifespan: a prospective study. Lancet Respir Med. 2015;3(8):613–620. doi: 10.1016/S2213-2600(15)00196-4.
    1. Bernard A, Marchandise FX, Depelchin S, Lauwerys R, Sibille Y. Clara cell protein in serum and bronchoalveolar lavage. Eur Respir J. 1992;5(10):1231–1238.
    1. Broeckaert F, Clippe A, Knoops B, Hermans C, Bernard A. Clara cell secretory protein (CC16): features as a peripheral lung biomarker. Ann N Y Acad Sci. 2000;923:68–77. doi: 10.1111/j.1749-6632.2000.tb05520.x.
    1. Laucho-Contreras ME, Polverino F, Gupta K, Taylor KL, Kelly E, Pinto-Plata V, Divo M, Ashfaq N, Petersen H, Stripp B, et al. Protective role for club cell secretory protein-16 (CC16) in the development of COPD. Eur Respir J. 2015;45(6):1544–1556. doi: 10.1183/09031936.00134214.
    1. Zhu L, Di PY, Wu R, Pinkerton KE, Chen Y. Repression of CC16 by cigarette smoke (CS) exposure. PLoS One. 2015;10(1):e0116159. doi: 10.1371/journal.pone.0116159.
    1. Vestbo J, Edwards LD, Scanlon PD, Yates JC, Agusti A, Bakke P, Calverley PM, Celli B, Coxson HO, Crim C, et al. Changes in forced expiratory volume in 1 second over time in COPD. N Engl J Med. 2011;365(13):1184–1192. doi: 10.1056/NEJMoa1105482.
    1. Park HY, Churg A, Wright JL, Li Y, Tam S, Man SF, Tashkin D, Wise RA, Connett JE, Sin DD. Club cell protein 16 and disease progression in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;188(12):1413–1419. doi: 10.1164/rccm.201305-0892OC.
    1. Lomas DA, Silverman EK, Edwards LD, Miller BE, Coxson HO, Tal-Singer R, Evaluation of CLtIPSEi: Evaluation of serum CC-16 as a biomarker for COPD in the ECLIPSE cohort. Thorax 2008, 63(12):1058–1063.
    1. Petersen H, Leng S, Belinsky SA, Miller BE, Tal-Singer R, Owen CA, Celli B, Tesfaigzi Y. Low plasma CC16 levels in smokers are associated with a higher risk for chronic bronchitis. Eur Respir J. 2015;46(5):1501–1503. doi: 10.1183/13993003.00682-2015.
    1. Laucho-Contreras ME, Polverino F, Tesfaigzi Y, Pilon A, Celli BR, Owen CA. Club cell protein 16 (CC16) augmentation: a potential disease-modifying approach for chronic obstructive pulmonary disease (COPD). Expert Opin Ther Targets. 2016;20(7):869–83.
    1. Ip MS, Ko FW, Lau AC, Yu WC, Tang KS, Choo K, Chan-Yeung MM. Hong Kong thoracic S, American College of Chest P: updated spirometric reference values for adult Chinese in Hong Kong and implications on clinical utilization. Chest. 2006;129(2):384–392. doi: 10.1378/chest.129.2.384.
    1. Lau AC, Ip MS, Lai CK, Choo KL, Tang KS, Yam LY, Chan-Yeung M. Variability of the prevalence of undiagnosed airflow obstruction in smokers using different diagnostic criteria. Chest. 2008;133(1):42–48. doi: 10.1378/chest.07-1434.
    1. Miller MR, Hankinson J, Brusasco V, Burgos F, Casaburi R, Coates A, Crapo R, Enright P, van der Grinten CP, Gustafsson P, et al. Standardisation of spirometry. Eur Respir J. 2005;26(2):319–338. doi: 10.1183/09031936.05.00034805.
    1. Ramirez RD, Sheridan S, Girard L, Sato M, Kim Y, Pollack J, Peyton M, Zou Y, Kurie JM, Dimaio JM, et al. Immortalization of human bronchial epithelial cells in the absence of viral oncoproteins. Cancer Res. 2004;64(24):9027–9034. doi: 10.1158/0008-5472.CAN-04-3703.
    1. Lam DC, Luo SY, Fu KH, Lui MM, Chan KH, Wistuba GB, II, Tsao SW, Ip MS, Minna JD. Nicotinic acetylcholine receptor expression in human airway correlates with lung function. Am J Physiol Lung Cell Mol Physiol. 2016;310(3):L232–L239. doi: 10.1152/ajplung.00101.2015.
    1. Lau WK, Chan SC, Law AC, Ip MS, Mak JC. The role of MAPK and Nrf2 pathways in ketanserin-elicited attenuation of cigarette smoke-induced IL-8 production in human bronchial epithelial cells. Toxicol Sci. 2012;125(2):569–577. doi: 10.1093/toxsci/kfr305.
    1. Dye BR, Hill DR, Ferguson MA, Tsai YH, Nagy MS, Dyal R, Wells JM, Mayhew CN, Nattiv R, Klein OD, et al. In vitro generation of human pluripotent stem cell derived lung organoids. elife. 2015;4.
    1. Glushakova O, Kosugi T, Roncal C, Mu W, Heinig M, Cirillo P, Sanchez-Lozada LG, Johnson RJ, Nakagawa T. Fructose induces the inflammatory molecule ICAM-1 in endothelial cells. J Am Soc Nephrol. 2008;19(9):1712–1720. doi: 10.1681/ASN.2007121304.
    1. Yao XL, Ikezono T, Cowan M, Logun C, Angus CW, Shelhamer JH. Interferon-gamma stimulates human Clara cell secretory protein production by human airway epithelial cells. Am J Phys. 1998;274(5 Pt 1):L864–L869.
    1. Cowan MJ, Huang X, Yao XL, Shelhamer JH. Tumor necrosis factor alpha stimulation of human Clara cell secretory protein production by human airway epithelial cells. Ann N Y Acad Sci. 2000;923:193–201. doi: 10.1111/j.1749-6632.2000.tb05530.x.
    1. Yoon JM, Lee KH, Lee SM, Lim JJ, Yang SC, Yoo CG, Lee CT, Han SK, Shim YS, Kim YW. The immune modulation of Clara cell-10 in human peripheral monocytes and dendritic cells. Int J Mol Med. 2010;26(3):415–423.

Source: PubMed

3
S'abonner