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

Mingxing Yang, Maxie Kohler, Tina Heyder, Helena Forsslund, Hilde K Garberg, Reza Karimi, Johan Grunewald, Frode S Berven, C Magnus Sköld, Åsa M Wheelock, Mingxing Yang, Maxie Kohler, Tina Heyder, Helena Forsslund, Hilde K Garberg, Reza Karimi, Johan Grunewald, Frode S Berven, C Magnus Sköld, Åsa M Wheelock

Abstract

Background: Smoking represents a significant risk factor for many chronic inflammatory diseases, including chronic obstructive pulmonary disease (COPD).

Methods: To identify dysregulation of specific proteins and pathways in bronchoalveolar lavage (BAL) cells associated with smoking, isobaric tags for relative and absolute quantitation (iTRAQ)-based shotgun proteomics analyses were performed on BAL cells from healthy never-smokers and smokers with normal lung function from the Karolinska COSMIC cohort. Multivariate statistical modeling, multivariate correlations with clinical data, and pathway enrichment analysis were performed.

Results: Smoking exerted a significant impact on the BAL cell proteome, with more than 500 proteins representing 15 molecular pathways altered due to smoking. The majority of these alterations occurred in a gender-independent manner. The phagosomal- and leukocyte trans endothelial migration (LTM) pathways significantly correlated with FEV1/FVC as well as the percentage of CD8+ T-cells and CD8+CD69+ T-cells in smokers. The correlations to clinical parameters in healthy never-smokers were minor.

Conclusion: The significant correlations of proteins in the phagosome- and LTM pathways with activated cytotoxic T-cells (CD69+) and the level of airway obstruction (FEV1/FVC) in smokers, both hallmarks of COPD, suggests that these two pathways may play a role in the molecular events preceding the development of COPD in susceptible smokers. Both pathways were found to be further dysregulated in COPD patients from the same cohort, thereby providing further support to this hypothesis. Given that not all smokers develop COPD in spite of decades of smoking, it is also plausible that some of the molecular pathways associated with response to smoking exert protective mechanisms to smoking-related pathologies in resilient individuals.

Trial registration: ClinicalTrials.gov identifier NCT02627872 ; Retrospectively registered on December 9, 2015.

Keywords: Bronchoalveolar lavage; COPD; Inflammation; Proteomics; Smoking.

Conflict of interest statement

Ethics approval and consent to participate

The study was approved by the Stockholm regional ethical board (Case no. 2006/959–31/1), and written informed consent was obtained from all subjects.

Consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

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

Figures

Fig. 1
Fig. 1
Flow chart of the study design outlining groups of the current paper as well as the companion manuscript [15]. A total of 69 subjects from the Karolinska COSMIC cohort, age- and gender- matched, were selected for iTRAQ proteomic investigations, including 18 healthy Never-smokers, 25 Smokers with normal lung function, 18 current smokers with COPD, and 8 ex-smokers with COPD. The current analysis consists of 17 Never-smokers (one male Never-smoker did not pass QC and was excluded from further analysis), 25 Smokers with normal lung function. This report focuses on the alterations in proteomes and pathways related to smokingwhereas the pathways related to pathogenesis of COPD are reported in a companion paper [15]
Fig. 2
Fig. 2
a) Scores plots of OPLS-DA modeling of BAL cell proteome alterations between Smokers (circle) vs Never-smokers (open triangles) OPLS-DA models suggested a perfect separation and predictive power between Smokers and Never-smokers p(CV-ANOVA) = 6.2 × 10−19) using 506 significant proteins (|p(corr)| > 0.34, n = 42, Additional file 1: Table S2). Stratification by gender revealed a highly significant separation in females (b; p(CV-ANOVA) = 3.2 × 10−10), fitted using 401 significant proteins (Additional file 1: Table S3), with a near perfect predictive power of 98% (R2 = 0.98, Q2 = 0.94). Also in males, the group separation was significant (c; p(CV-ANOVA) = 4.5 × 10−6), fitted with 301 significant proteins (Additional file 1: Table S4), with a predictive power of 87% (R2 = 0.95, Q2 = 0.87) . As displayed by the Venn diagram in panel d, a total of 199 proteins were altered in both female and male Smokers, while 202 proteins were altered only in female Smokers, and 102 proteins were altered only in male Smokers. An additional 75 proteins were found to be altered only in the joint gender model. The majority of the significantly altered proteins from the gender stratified models were altered also in the joint gender model; 95% in male and 86% female Smokers. Keys: t[1]: scores of OPLS-DA predictive component; to[1]: scores of the first orthogonal component from OPLS-DA model; p:cross-validated (CV)-ANOVA p-value for significance of group separation in the model
Fig. 3
Fig. 3
The phagosomal pathway was enriched in Smokers vs Never-smokers in the joint gender model (p = 1.4 × 10−6, FDR = 2.7 × 10−5). Proteins with significantly increased levels are highlighted in red, and decreased levels in dark blue. (FcγR, P08637, Low affinity immunoglobulin γ Fc region receptor III-A; CR3, P11215/P05107, CD11b/CD18; αMβ2, P11215/P05107, CD11b/CD18; Collectins, Q8IWL1, Pulmonary surfactant-associated protein A2; SRA1, P21757, Macrophage scavenger receptor types I and II; MARCO, Q9UEW3, Macrophage receptor MARCO; TAP1, Q03518, Antigen peptide transporter 1; TAP2, Q03519, Antigen peptide transporter 2; F-actin, P60709, β-actin; Stx13, Q86Y82, Syntaxin-12; TfR, P02786, Transferrin receptor protein 1; LAMP, P13473, lysosome-associated membrane glycoprotein 2; Cathepsin S: P25774, Cathepsin S; Cathepsin L1: P07711, Cathepsin L1; vATPase: P38606, P21281, P61421, P21283, Q9Y5K8, P36543, O75348, Q9UI12, V-ATPase subunit A, B2, d1, C1, D, E1, G1, H, respectively; gp91, P04839, Cytochrome b-245 heavy chain; p40phox, Q15080, Neutrophil cytosol factor 4. Protein details are listed in Additional file 1: Table S5)
Fig. 4
Fig. 4
Gender-specific alterations in glycolysis/gluconeogenesis in Smokers. a The increased expression levels (in red) of five proteins (HK2, FBP1, FBP2, ALDOA and ENO1) in the female Smoker. b The decreased levels (in blue) of four proteins (HK2, PGK1, PGH2 and LDHA) in male Smokers. The levels of four proteins (DLAT, PDHA1, DLD and PDHB) in pathway pyruvate metabolism were increased in both gender Smokers
Fig. 5
Fig. 5
Pathways altered both due to smoking (left panels) and COPD (right panels) in females. The majority of expression levels in oxidative phosphorylation increased in the female Smoker compared to Never-smoker groups (a), and were further elevated in female current-smoker COPD compared with female Smoker groups (b), with six shared proteins (S). The levels of proteins in the citrate cycle were elevated in female Smokers compared to Never-smokers (c), and further increased in female smokers with COPD (d), with 4 proteins in common (S). The majority of protein levels in the lysosomal pathway were increased in female Smokers (e), but decreased in female smokers with COPD (f), with one common protein altered in the opposite direction (S). The bars display the scaled loadings, p(corr) of the predictive component from OPLS-DA models. Positive p(corr)[1] indicates increased protein levels. Full protein names are provided in Additional file 1: Table S3
Fig. 6
Fig. 6
The proportion of CD8+ T cells in BAL increased and CD4+ T-cells decreased in Smokers compared to Never-smokers (a). Proteins from the leukocyte transendothelial migration (LTM) pathway and phagosome pathway correlated with proportion of CD8+ T-cells (b-c), as well as with the proportion of CD8+ CD69+ T-cells (e-f). Protein levels from the LTM pathway also correlated with the level of lung obstruction; FEV1/FVC (d). R2 and p-values refer to PLS inner relation
Fig. 7
Fig. 7
Comparison of the proteomes investigated with the bottom-up iTRAQ peptidomics platform and the top-down 2D–DIGE intact proteomics platform. a) Venn diagram displaying the overlap of proteins identified with the two platforms indicates that the majority of proteins identified with iTRAQ (90%) were novel as compared to 2D–DIGE. The proteins that overlapped were generally altered in a similar fashion, thereby providing validation across platforms. b) Protein 60S acidic ribosomal protein P0 with decreased level in ribosomal pathway in Smokers (2-D DIGE: p = 5.1 × 10−8; iTRAQ: p = 0.009); c) Protein Cytochrome b-c1 complex subunit Rieske with elevated level in pathway oxidative phosphorylation in Smokers (2-D DIGE: p = 4.5 × 10−9; iTRAQ: p = 1.5 × 10−5); d) Protein Cathepsin D with increased level in lysosomal pathway in Smokers (2D DIGE: p = 4.0 × 10−17; iTRAQ: p = 4.3 × 10−19); e) ATP synthase subunit d in mitochondria with upregulated level in citrate cycle in Smokers (2-D DIGE: p = 4.0 × 10−5; iTRAQ: p = 6.3 × 10−5); f) Enzyme Aldehyde dehydrogenase in mitochondria with downregulated level in pathway valine, leucine and isoleucine degradation in Smokers (2-D DIGE: p = 8.4 × 10−8; iTRAQ: p = 0.02); g) Protein actin, cytoplasmic 1 with decreased level in pathway phagosome in smokers (2-D DIGE: p = 4.4 × 10−6; iTRAQ: p = 0.007)

References

    1. Ng M, Freeman MK, Fleming TD, et al. Smoking prevalence and cigarette consumption in 187 countries, 1980-2012. JAMA. 2014;311(2):183–192. doi: 10.1001/jama.2013.284692.
    1. Mathers CD, Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 2006;3(11):e442. doi: 10.1371/journal.pmed.0030442.
    1. Crotty Alexander LE, Shin S, Hwang JH. Inflammatory diseases of the lung induced by conventional cigarette smoke: a review. Chest. 2015;148(5):1307–1322. doi: 10.1378/chest.15-0409.
    1. Bortner JD, Jr., Richie JP, Jr., Das A, et al. Proteomic profiling of human plasma by iTRAQ reveals down-regulation of ITI-HC3 and VDBP by cigarette smoking. J Proteome Res 2011;10(3):1151-1159.
    1. Airoldi L, Magagnotti C, Iannuzzi AR, et al. Effects of cigarette smoking on the human urinary proteome. Biochem Biophys Res Commun. 2009;381(3):397–402. doi: 10.1016/j.bbrc.2009.02.055.
    1. Kelsen SG, Duan X, Ji R, Perez O, Liu C, Merali S. Cigarette smoke induces an unfolded protein response in the human lung: a proteomic approach. Am J Respir Cell Mol Biol. 2008;38(5):541–550. doi: 10.1165/rcmb.2007-0221OC.
    1. Sopori M. Effects of cigarette smoke on the immune system. Nat Rev Immunol. 2002;2(5):372–377. doi: 10.1038/nri803.
    1. Kohler M, Sandberg A, Kjellqvist 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. Forsslund H, Mikko M, Karimi R, 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. Karimi R, Tornling G, Forsslund H, 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. Forsslund H, Yang M, Mikko M, 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. Balgoma D, Yang M, Sjodin M, 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. Karimi R, Tornling G, Forsslund H, et al. Differences in regional air trapping in current smokers with normal spirometry. Eur Respir J. 2017;49(1).
    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. Yang M, Kohler M, Heyder T, Forsslund H, Garberg HK, Grunewald J, et al. Proteomic profiling of lung immune cells reveals dysregulation of phagocytotic pathways in female-dominated molecular COPD phenotype. Respir Res. 2018;19(1). 10.1186/s12931-017-0699-2.
    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. Storey JD. A direct approach to false discovery rates. J Royal Stat Soc. 2002;64(3):479–498. doi: 10.1111/1467-9868.00346.
    1. Bylesjö M, Rantalainen M, Cloarec O, Nicholson JK, Holmes E, Trygg J. OPLS discriminant analysis: combining the strengths of PLS-DA and SIMCA classification. J Chemom. 2006;20(8–10):341–351. doi: 10.1002/cem.1006.
    1. Wheelock AM, Wheelock CE. Trials and tribulations of ‘omics data analysis: assessing quality of SIMCA-based multivariate models using examples from pulmonary medicine. Mol BioSyst. 2013;9(11):2589–2596. doi: 10.1039/c3mb70194h.
    1. Eriksson L, Trygg J, Wold S. CV-ANOVA for significance testing of PLS and OPLS® models. J Chemom. 2008;22(11–12):594–600. doi: 10.1002/cem.1187.
    1. Levanen B, Bhakta NR, Torregrosa Paredes P, et al. Altered microRNA profiles in bronchoalveolar lavage fluid exosomes in asthmatic patients. J Allergy Clin Immunol. 2013;131(3):894–903. doi: 10.1016/j.jaci.2012.11.039.
    1. Xie C, Mao X, Huang J, et al. KOBAS 2.0: a web server for annotation and identification of enriched pathways and diseases. Nucleic Acids Res. 2011;39(Web Server issue):W316–W322. doi: 10.1093/nar/gkr483.
    1. Kanehisa M, Goto S, Sato Y, Furumichi M, Tanabe M. KEGG for integration and interpretation of large-scale molecular data sets. Nucleic Acids Res. 2012;40(Database issue):D109–D114. doi: 10.1093/nar/gkr988.
    1. Maeno T, Houghton AM, Quintero PA, Grumelli S, Owen CA, Shapiro SD. CD8+ T cells are required for inflammation and destruction in cigarette smoke-induced emphysema in mice. J Immunol. 2007;178(12):8090–8096. doi: 10.4049/jimmunol.178.12.8090.
    1. Freeman CM, Han MK, Martinez FJ, et al. Cytotoxic potential of lung CD8(+) T cells increases with chronic obstructive pulmonary disease severity and with in vitro stimulation by IL-18 or IL-15. J Immunol. 2010;184(11):6504–6513. doi: 10.4049/jimmunol.1000006.
    1. Chung KF, Adcock IM. Multifaceted mechanisms in COPD: inflammation, immunity, and tissue repair and destruction. Eur Respir J. 2008;31(6):1334–1356. doi: 10.1183/09031936.00018908.
    1. Gonzalez-Amaro R, Cortes JR, Sanchez-Madrid F, Martin P. Is CD69 an effective brake to control inflammatory diseases? Trends Mol Med. 2013;19(10):625–632. doi: 10.1016/j.molmed.2013.07.006.
    1. Tsuyusaki J, Kuroda F, Kasuya Y, et al. Cigarette smoke-induced pulmonary inflammation is attenuated in CD69-deficient mice. J Recept Signal Transduct Res. 2011;31(6):434–439. doi: 10.3109/10799893.2011.631929.
    1. Fujita T, Yoshioka K, Umezawa H, et al. Role of CD69 in the pathogenesis of elastase-induced pulmonary inflammation and emphysema. Biochem Biophys Rep. 2016;7:400–407.
    1. Haas A. The phagosome: compartment with a license to kill. Traffic. 2007;8(4):311–330. doi: 10.1111/j.1600-0854.2006.00531.x.
    1. Fairn GD, Grinstein S. How nascent phagosomes mature to become phagolysosomes. Trends Immunol. 2012;33(8):397–405. doi: 10.1016/j.it.2012.03.003.
    1. Collins RF, Schreiber AD, Grinstein S, Trimble WS. Syntaxins 13 and 7 function at distinct steps during phagocytosis. J Immunol. 2002;169(6):3250–3256. doi: 10.4049/jimmunol.169.6.3250.
    1. Huynh KK, Eskelinen EL, Scott CC, Malevanets A, Saftig P, Grinstein S. LAMP proteins are required for fusion of lysosomes with phagosomes. EMBO J. 2007;26(2):313–324. doi: 10.1038/sj.emboj.7601511.
    1. Kinchen JM, Ravichandran KS. Phagosome maturation: going through the acid test. Nat Rev Mol Cell Biol. 2008;9(10):781–795. doi: 10.1038/nrm2515.
    1. Hodge S, Hodge G, Ahern J, Jersmann H, Holmes M, Reynolds PN. Smoking alters alveolar macrophage recognition and phagocytic ability: implications in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2007;37(6):748–755. doi: 10.1165/rcmb.2007-0025OC.
    1. Kennedy DW, Abkowitz JL. Mature monocytic cells enter tissues and engraft. Proc Natl Acad Sci U S A. 1998;95(25):14944–14949. doi: 10.1073/pnas.95.25.14944.
    1. Muller WA. Mechanisms of leukocyte transendothelial migration. Annu Rev Pathol. 2011;6:323–344. doi: 10.1146/annurev-pathol-011110-130224.
    1. Mamdouh Z, Chen X, Pierini LM, Maxfield FR, Muller WA. Targeted recycling of PECAM from endothelial surface-connected compartments during diapedesis. Nature. 2003;421(6924):748–753. doi: 10.1038/nature01300.
    1. Muller WAPECAM. Regulating the start of diapedesis. In: Ley K, editor. Adhesion Molecules: Function and Inhibition. Basel: Birkhäuser Basel; 2007. pp. 201–220.
    1. Blidberg K, Palmberg L, James A, et al. Adhesion molecules in subjects with COPD and healthy non-smokers: a cross sectional parallel group study. Respir Res. 2013;14:47. doi: 10.1186/1465-9921-14-47.
    1. Solovjov DA, Pluskota E, Plow EF. Distinct roles for the alpha and beta subunits in the functions of integrin alphaMbeta2. J Biol Chem. 2005;280(2):1336–1345. doi: 10.1074/jbc.M406968200.
    1. Kirby AC, Raynes JG, Kaye PM. CD11b regulates recruitment of alveolar macrophages but not pulmonary dendritic cells after pneumococcal challenge. J Infect Dis. 2006;193(2):205–213. doi: 10.1086/498874.
    1. Amaral AFS, Strachan DP, Burney PGJ, Jarvis DL. Female smokers are at greater risk of airflow obstruction than male smokers. UK biobank. Am J Respir Crit Care Med. 2017;195(9):1226–1235. doi: 10.1164/rccm.201608-1545OC.

Source: PubMed

3
Předplatit