Gender differences in the T-cell profiles of the airways in COPD patients associated with clinical phenotypes

Helena Forsslund, Mingxing Yang, Mikael Mikko, Reza Karimi, Sven Nyrén, Benita Engvall, Johan Grunewald, Heta Merikallio, Riitta Kaarteenaho, Jan Wahlström, Åsa M Wheelock, C Magnus Sköld, Helena Forsslund, Mingxing Yang, Mikael Mikko, Reza Karimi, Sven Nyrén, Benita Engvall, Johan Grunewald, Heta Merikallio, Riitta Kaarteenaho, Jan Wahlström, Åsa M Wheelock, C Magnus Sköld

Abstract

T lymphocytes are believed to play an important role in the pathogenesis of chronic obstructive pulmonary disease (COPD). How T cells are recruited to the lungs and contribute to the inflammatory process is largely unknown. COPD is a heterogeneous disease, and discriminating disease phenotypes based on distinct molecular and cellular pathways may provide new approaches for individualized diagnosis and therapies. Bronchoalveolar lavage (BAL) and blood samples were obtained from 40 never-smokers, 40 smokers with normal lung function, and 38 COPD patients. T-cell chemokine receptor expression was analyzed with flow cytometry, and soluble BAL cytokines and chemokines were measured using a cytokine multiplex assay. Correlations with gender and clinical characteristics including lung imaging were investigated using multivariate modeling. Th1/Tc1- and Th2/Tc2-associated soluble analytes and T-cell chemokine receptors were analyzed as cumulative Th1/Tc1 and Th2/Tc2 immune responses. A higher expression of chemokine receptor CCR5 on CD8+ T cells in BAL and higher percentage of CXCR3+CD8+ T cells in blood was found in female smokers with COPD compared to those without COPD. CCR5 expression on CD4+ and CD8+ T cells was lower in BAL from male smokers with COPD compared to those without COPD. Among female smokers with COPD, Th1/Tc1 immune response was linked to BAL macrophage numbers and goblet cell density, and Th2/Tc2 response was associated with the measures of emphysema on high-resolution computed tomography. The highly gender-dependent T-cell profile in COPD indicates different links between cellular events and clinical manifestations in females compared to males. Our findings may reveal mechanisms of importance for the difference in clinical course in female COPD patients compared to males.

Keywords: Th1/Th2; bronchoalveolar lavage; chemokines; cytokines.

Conflict of interest statement

The authors report no conflicts of interest in this work.

Figures

Figure 1
Figure 1
Results of univariate analysis. Notes: Percentages of CXCR3+ among (A) CD4+ and (B) CD8+ T cells in BAL. MFI of CCR5 on (C) CD4+ and (D) CD8+ T cells and the MFI of CXCR4 on (E) CD4+ and (F) CD8+ T cells in BAL from healthy never-smokers (NS), smokers with normal lung function (S), smokers with COPD (CS), and ex-smokers with COPD (CE). Horizontal lines represent the median values. **P<0.01. Abbreviations: BAL, bronchoalveolar lavage; COPD, chronic obstructive pulmonary disease; MFI, median fluorescence intensity.
Figure 2
Figure 2
Relative concentrations of (A) IL-13 and (B) CXCL12 in BAL supernatant from healthy never-smokers (NS), smokers with normal lung function (S), smokers with COPD (CS), and ex-smokers with COPD (CE). Notes: Data have been group mean-centered for the removal of batch effects. Horizontal lines represent the median values. *P<0.05; **P<0.01; ***P<0.001. Abbreviations: BAL, bronchoalveolar lavage; IL, interleukin.
Figure 3
Figure 3
Multivariate modeling comparing T-cell subsets and soluble analytes of smokers with normal lung function versus COPD smokers. Notes: (A) OPLS-DA resulted in a significant separation between female smokers and female COPD patients (R2=0.50, Q2=0.41, P=8×10−4). Left panel shows the score plot with the predictive component along the x-axis and the y-axis indicating the numerical order of the subjects. The right panel shows the loadings plot, with primarily eleven variables driving a separation. (B) Among males, the analysis resulted in a significant separation between male smokers and male COPD patients (R2=0.32, Q2=0.25, P=0.016) with primarily eight variables driving a separation. Boxes above zero are markers upregulated and boxes below zero are markers downregulated due to COPD. Black dots: COPD smokers; white dots: smokers with normal lung function. Abbreviations: BAL, bronchoalveolar lavage; COPD, chronic obstructive pulmonary disease; IFN, interferon; IL, interleukin; MFI, median fluorescence intensity; OPLS-DA, orthogonal projections to latent structures discriminate analysis; TGF, transforming growth factor.
Figure 4
Figure 4
Results of univariate analysis on gender-associated factors separate in smokers with normal lung function and COPD smokers. Notes: MFI of CCR5 on CD8+ T cells in (A) blood and (B) BAL. (C) Percentage of CXCR3+ cells among CD8+ T cells in blood, from female smokers and female COPD smokers. MFI of CCR5 on both (D) CD4+ T cells and (E) CD8+ T cells in BAL from male smokers and male COPD smokers. Horizontal lines represent the median values.*P<0.05, **P<0.01. Abbreviations: BAL, bronchoalveolar lavage; COPD, chronic obstructive pulmonary disease; MFI, median fluorescence intensity.
Figure 5
Figure 5
SUS analysis comparing COPD-specific cytokine and T-cell profiles in the male (y-axis) versus female (x-axis) smoking populations. Notes: Variables along the negative diagonal indicate the opposite response to COPD in males and females (variables 1–5), while variables along the outer edges of the y-axis indicate a response to COPD in females, but not males (variable 6). P(corr) [1], the scaled loadings of predictive component for the respective OPLS model. Abbreviations: BAL, bronchoalveolar lavage; COPD, chronic obstructive pulmonary disease; OPLS, orthogonal projections to latent structures; SUS, Shared and Unique Structures.
Figure 6
Figure 6
Associations between pathways and clinical parameters. Notes: Significant associations between pathways and clinical parameters were found among female COPD smokers for the (A) Th2/Tc2 pathway and HRCT attenuation <−950 HU (P=0.002), (B) the Th1/Tc1 pathway and macrophage numbers in BAL (P=0.006) as well as with (C) lung airway goblet cell density (P=0.02). Among male smokers, a significant association was found between the (D) Th2/Tc2 pathway and serum IgG concentrations (P=0.003). Values of r represent PLS inner relation between the respective clinical variables and the respective analytes involved in the respective pathway. Th1/Tc1 pathway: CCR5, CXCR3, CD69 receptors, and BAL mediators TNF-α, IFN-γ, CCL3–5, CXCL9–10. Th2/Tc2 pathway: CXCR4, CD69 receptors and BAL mediators IL-4, IL-13, and CXCL12. u, latent scores variable for the Y-block of PLS; t, latent scores variable for the X-block of PLS. Abbreviations: BAL, bronchoalveolar lavage; COPD, chronic obstructive pulmonary disease; HRCT, high-resolution computed tomography; HU, Hounsfield units; IFN, interferon; IL, interleukin; PLS, partial least squares; TNF, tumor necrosis factor.

References

    1. The National Board of Health and Welfare (Socialstyrelsen) Hälsooch sjukvårdsrapport 2009. [Accessed October 6, 2016]. Socialstyrelsen; Stockholm, Sweden. Available from: .
    1. Mannino DM, Homa DM, Akinbami LJ, Ford ES, Redd SC. Chronic obstructive pulmonary disease surveillance – United States, 1971–2000. MMWR Surveill Summ. 2002;51(6):1–16.
    1. Akinbami LJ, Liu X. Chronic obstructive pulmonary disease among adults aged 18 and over in the United States, 1998–2009. NCHS Data Brief. 2011;63:1–8.
    1. Langhammer A, Johnsen R, Gulsvik A, Holmen TL, Bjermer L. Sex differences in lung vulnerability to tobacco smoking. Eur Respir J. 2003;21(6):1017–1023.
    1. Ben-Zaken Cohen S, Pare PD, Man SF, Sin DD. The growing burden of chronic obstructive pulmonary disease and lung cancer in women: examining sex differences in cigarette smoke metabolism. Am J Respir Crit Care Med. 2007;176(2):113–120.
    1. Brusselle GG, Joos GF, Bracke KR. New insights into the immunology of chronic obstructive pulmonary disease. Lancet. 2011;378(9795):1015–1026.
    1. Hogg JC, Timens W. The pathology of chronic obstructive pulmonary disease. Annu Rev Pathol. 2009;4:435–459.
    1. Enelow RI, Mohammed AZ, Stoler MH, et al. Structural and functional consequences of alveolar cell recognition by CD8(+) T lymphocytes in experimental lung disease. J Clin Invest. 1998;102(9):1653–1661.
    1. Finkelstein R, Fraser RS, Ghezzo H, Cosio MG. Alveolar inflammation and its relation to emphysema in smokers. Am J Respir Crit Care Med. 1995;152(5 Pt 1):1666–1672.
    1. Freeman CM, Curtis JL, Chensue SW. CC chemokine receptor 5 and CXC chemokine receptor 6 expression by lung CD8+ cells correlates with chronic obstructive pulmonary disease severity. Am J Pathol. 2007;171(3):767–776.
    1. Barcelo B, Pons J, Fuster A, et al. Intracellular cytokine profile of T lymphocytes in patients with chronic obstructive pulmonary disease. Clin Exp Immunol. 2006;145(3):474–479.
    1. Barczyk A, Pierzchala W, Kon OM, Cosio B, Adcock IM, Barnes PJ. Cytokine production by bronchoalveolar lavage T lymphocytes in chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2006;117(6):1484–1492.
    1. Syrbe U, Siveke J, Hamann A. Th1/Th2 subsets: distinct differences in homing and chemokine receptor expression? Springer Semin Immunopathol. 1999;21(3):263–285.
    1. Onuffer JJ, Horuk R. Chemokines, chemokine receptors and small-molecule antagonists: recent developments. Trends Pharmacol Sci. 2002;23(10):459–467.
    1. Palmer LA, Sale GE, Balogun JI, et al. Chemokine receptor CCR5 mediates alloimmune responses in graft-versus-host disease. Biol Blood Marrow Transplant. 2010;16(3):311–319.
    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.
    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.
    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.
    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.
    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.
    1. Bancroft JD, Gamble M. Theory and Practice of Histological Techniques. 6th ed. Edinburgh: Churchill Livingstone; 2008.
    1. Wong DM, Varesio L. Depletion of macrophages from heterogeneous cell populations by the use of carbonyl iron. Methods Enzymol. 1984;108:307–313.
    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.
    1. Gevenois PA, De Vuyst P, de Maertelaer V, et al. Comparison of computed density and microscopic morphometry in pulmonary emphysema. Am J Respir Crit Care Med. 1996;154(1):187–192.
    1. Saetta M, Mariani M, Panina-Bordignon P, et al. Increased expression of the chemokine receptor CXCR3 and its ligand CXCL10 in peripheral airways of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2002;165(10):1404–1409.
    1. Ohar J, Fromer L, Donohue JF. Reconsidering sex-based stereotypes of COPD. Prim Care Respir J. 2011;20(4):370–378.
    1. Kohlmeier JE, Miller SC, Smith J, et al. The chemokine receptor CCR5 plays a key role in the early memory CD8+ T cell response to respiratory virus infections. Immunity. 2008;29(1):101–113.
    1. Fukada K, Sobao Y, Tomiyama H, Oka S, Takiguchi M. Functional expression of the chemokine receptor CCR5 on virus epitope-specific memory and effector CD8+ T cells. J Immunol. 2002;168(5):2225–2232.
    1. Packard TA, Li QZ, Cosgrove GP, Bowler RP, Cambier JC. COPD is associated with production of autoantibodies to a broad spectrum of self-antigens, correlative with disease phenotype. Immunol Res. 2013;55(1–3):48–57.
    1. Daffa NI, Tighe PJ, Corne JM, Fairclough LC, Todd I. Natural and disease-specific autoantibodies in chronic obstructive pulmonary disease. Clin Exp Immunol. 2015;180(1):155–163.
    1. Selmi C, Brunetta E, Raimondo MG, Meroni PL. The X chromosome and the sex ratio of autoimmunity. Autoimmun Rev. 2012;11(6–7):A531–A537.
    1. Tam A, Morrish D, Wadsworth S, Dorscheid D, Man SF, Sin DD. The role of female hormones on lung function in chronic lung diseases. BMC Womens Health. 2011;11:24.
    1. Tai P, Wang J, Jin H, et al. Induction of regulatory T cells by physiological level estrogen. J Cell Physiol. 2008;214(2):456–464.
    1. Grumelli S, Corry DB, Song LZ, et al. An immune basis for lung parenchymal destruction in chronic obstructive pulmonary disease and emphysema. PLoS Med. 2004;1(1):e8.
    1. Shan M, Cheng HF, Song LZ, et al. Lung myeloid dendritic cells coordinately induce TH1 and TH17 responses in human emphysema. Sci Transl Med. 2009;1(4):4ra10.
    1. Wang Z, Zheng T, Zhu Z, et al. Interferon gamma induction of pulmonary emphysema in the adult murine lung. J Exp Med. 2000;192(11):1587–1600.
    1. Zheng T, Zhu Z, Wang Z, et al. Inducible targeting of IL-13 to the adult lung causes matrix metalloproteinase- and cathepsin-dependent emphysema. J Clin Invest. 2000;106(9):1081–1093.
    1. Barnes PJ. The cytokine network in asthma and chronic obstructive pulmonary disease. J Clin Invest. 2008;118(11):3546–3556.

Source: PubMed

3
Abonner