An immune basis for lung parenchymal destruction in chronic obstructive pulmonary disease and emphysema

Sandra Grumelli, David B Corry, Li-Zhen Song, Ling Song, Linda Green, Joseph Huh, Joan Hacken, Rafael Espada, Remzi Bag, Dorothy E Lewis, Farrah Kheradmand, Sandra Grumelli, David B Corry, Li-Zhen Song, Ling Song, Linda Green, Joseph Huh, Joan Hacken, Rafael Espada, Remzi Bag, Dorothy E Lewis, Farrah Kheradmand

Abstract

Background: Chronic obstructive pulmonary disease and emphysema are a frequent result of long-term smoking, but the exact mechanisms, specifically which types of cells are associated with the lung destruction, are unclear.

Methods and findings: We studied different subsets of lymphocytes taken from portions of human lungs removed surgically to find out which lymphocytes were the most frequent, which cell-surface markers these lymphocytes expressed, and whether the lymphocytes secreted any specific factors that could be associated with disease. We found that loss of lung function in patients with chronic obstructive pulmonary disease and emphysema was associated with a high percentage of CD4+ and CD8+ T lymphocytes that expressed chemokine receptors CCR5 and CXCR3 (both markers of T helper 1 cells), but not CCR3 or CCR4 (markers of T helper 2 cells). Lung lymphocytes in patients with chronic obstructive pulmonary disease and emphysema secrete more interferon gamma--often associated with T helper 1 cells--and interferon-inducible protein 10 and monokine induced by interferon, both of which bind to CXCR3 and are involved in attracting T helper 1 cells. In response to interferon-inducible protein 10 and monokine induced by interferon, but not interferon gamma, lung macrophages secreted macrophage metalloelastase (matrix metalloproteinase-12), a potent elastin-degrading enzyme that causes tissue destruction and which has been linked to emphysema.

Conclusions: These data suggest that Th1 lymphoctytes in the lungs of people with smoking-related damage drive progression of emphysema through CXCR3 ligands, interferon-inducible protein 10, and monokine induced by interferon.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Chemokine Receptor Expression on Peripheral…
Figure 1. Chemokine Receptor Expression on Peripheral Lung Lymphocytes
(A) Single color histograms showing expression of chemokine receptors CCR4, CCR5, and CXCR3 from representative control and emphysema participants. (B) Pooled data from all participants (control, n = 10; emphysema, n = 18) showing percent (median ± SD) of total lung lymphocytes expressing CCR4 and CCR5. (C) Pooled data from same participants showing percent (median ± SD) CCR5 expression on CD4 (top) and CD8 (middle) T cells, and CXCR3 expression on unfractionated T cells (bottom) from the same participant groups. (D) Analysis of total lung lymphocyte chemokine receptor (median ± SD) profiles among participants with emphysema. Participants had either (1) lung volume reduction surgery for emphysema (non-cancer, n = 8) or (2) lung resection for treatment of small peripheral cancer (n = 10). Participants showed similar inflammatory indices as determined by CCR5 expression. In (B) and (C), *, p < 0.001; †, p = 0.01; ‡, p = 0.02; ∫, p = 0.007 (Mann-Whitney test) for the comparison of emphysema and control groups.
Figure 2. Expression of CXCR3 in Lungs…
Figure 2. Expression of CXCR3 in Lungs of Control and Emphysematous Smoker Individuals
(A) Representative forward and side-scatter characteristics of whole lung cells from a participant with COPD and emphysema. Anti-CD11b PE-conjugated and anti-CD14 FITC-conjugated antibodies detect lung macrophages (middle), and histogram of mean fluorescence intensity showing anti-CXCR3-Cy5 and control antibodies (cIg) detects lung macrophages in the patient with emphysema. (B) Pooled data from control individuals without (n = 5) and with (n = 8) emphysema. Columns are median, bars represent SD. *, p = 0.009 (Mann-Whitney test) for the comparison of emphysema and control participants. (C) Negative association between CXCR3 expression on CD3+ T cells and FEV1 percentage predicted based on an R2 goodness-of-fit statistic of 0.27 (p = 0.0089, r = −0.52, n = 24).
Figure 3. IFN-γ, MIG, and IP-10 Production…
Figure 3. IFN-γ, MIG, and IP-10 Production by Isolated Lung Lymphocytes
(A–C) Lung lymphocytes from control individuals and participants with emphysema were cultured without additional stimulation for 3 or 4 d and assessed for secretion of (A) IFN-γ, (B) MIG, and (C) IP-10 (control, n = 8; emphysema, n = 12). Columns are median, bars represent SD. *, p= 0.007; †, p = 0.01; ‡, p = 0.02 for the comparison of emphysema and control participants. (D) The same cells from a representative ex-smoker individual with emphysema were either left unstimulated (No ST) or treated with PMA/ionomycin (PMA/I) for 24 h and assessed for surface CD8 and CD69 expression and the intracytoplasmic accumulation of IFN-γ by flow cytometry. (E) Production of IL-4 by lung lymphocytes. Lung lymphocytes from a representative ex-smoker individual with emphysema were cultured for 24 h with or without PMA/ionomycin stimulation (PMA/I) and assessed for intracytoplasmic IL-4 and IFN-γ accumulation by flow cytometry.
Figure 4. Regulation of MMP12 by Type…
Figure 4. Regulation of MMP12 by Type 1 Cytokines
(A) CD14+, lymphocyte-depleted lung leukocytes were cultured with and without the indicated amounts of recombinant human IP-10 and IFN-γ, and supernatants were assessed for the presence of MMP12 by Western blotting. (B) Fold increase relative to unstimulated of MMP12 mRNA from lung macrophages stimulated without () and with (+) 500 ng/ml of IP-10 in the presence or absence of a function-blocking antibody to CXCR3 as determined by real-time PCR. (C and D) Lung tissue from a participant with emphysema (C) shows strong immune staining for MMP12 localized to macrophages (arrows), and (D) shows lung tissue from a control participant without emphysema and with undetectable MMP12. The insets show a high-power view of lung macrophages staining positive (C) and negative (D) for MMP12 (×60) *, p = 0.04.

References

    1. Barnes PJ. Potential novel therapies for chronic obstructive pulmonary disease. Novartis Found Symp. 2001;234:255–267.
    1. Senior RM. Mechanisms of COPD: Conference summary. Chest. 2000;117:320S–323S.
    1. Hogg JC. Chronic obstructive pulmonary disease: An overview of pathology and pathogenesis. Novartis Found Symp. 2001;234:4–19.
    1. Corry DB, Grunig G, Hadeiba H, Kurup VP, Warnock ML, et al. Requirements for allergen-induced airway hyperreactivity in T and B cell-deficient mice. Mol Med. 1998;4:344–355.
    1. Grunig G, Warnock M, Wakil AE, Venkayya R, Brombacher F, et al. Requirement for IL-13 independently of IL-4 in experimental asthma. Science. 1998;282:2261–2263.
    1. Kay AB. T cells as orchestrators of the asthmatic response. Ciba Found Symp. 1997;206:56–67.
    1. Fahy JV, Corry DB, Boushey HA. Airway inflammation and remodeling in asthma. Curr Opin Pulm Med. 2000;6:15–20.
    1. Cosio MG, Guerassimov A. Chronic obstructive pulmonary disease. Inflammation of small airways and lung parenchyma. Am J Respir Crit Care Med. 1999;160:S21–S25.
    1. Di Stefano A, Capelli A, Lusuardi M, Caramori G, Balbo P, et al. Decreased T lymphocyte infiltration in bronchial biopsies of subjects with severe chronic obstructive pulmonary disease. Clin Exp Allergy. 2001;31:893–902.
    1. Turato G, Zuin R, Saetta M. Pathogenesis and pathology of COPD. Respiration. 2001;68:117–128.
    1. Saetta M, Di Stefano A, Turato G, Facchini FM, Corbino L, et al. CD8+ T-lymphocytes in peripheral airways of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157:822–826.
    1. Zheng T, Zhu Z, Wang Z, Homer RJ, Ma B, et al. Inducible targeting of IL-13 to the adult lung causes matrix metalloproteinase- and cathepsin-dependent emphysema. J Clin Invest. 2000;106:1081–1093.
    1. Anthonisen NR, Connett JE, Kiley JP, Altose MD, Bailey WC, et al. Effects of smoking intervention and the use of an inhaled anticholinergic bronchodilator on the rate of decline of FEV1. The Lung Health Study. JAMA. 1994;272:1497–1505.
    1. He JQ, Connett JE, Anthonisen NR, Sandford AJ. Polymorphisms in the IL13, IL13RA1, and IL4RA genes and rate of decline in lung function in smokers. Am J Respir Cell Mol Biol. 2003;28:379–385.
    1. Baggiolini M, Loetscher P. Chemokines in inflammation and immunity. Immunol Today. 2000;21:418–420.
    1. Sallusto F, Lanzavecchia A. Understanding dendritic cell and T-lymphocyte traffic through the analysis of chemokine receptor expression. Immunol Rev. 2000;177:134–140.
    1. Luster AD. The role of chemokines in linking innate and adaptive immunity. Curr Opin Immunol. 2002;14:129–135.
    1. Xie H, Lim YC, Luscinskas FW, Lichtman AH. Acquisition of selectin binding and peripheral homing properties by CD4(+) and CD8(+) T cells. J Exp Med. 1999;189:1765–1776.
    1. Luther SA, Cyster JG. Chemokines as regulators of T cell differentiation. Nat Immunol. 2001;2:102–107.
    1. Kim CH, Rott L, Kunkel EJ, Genovese MC, Andrew DP, et al. Rules of chemokine receptor association with T cell polarization in vivo. J Clin Invest. 2001;108:1331–1339.
    1. Loetscher P, Uguccioni M, Bordoli L, Baggiolini M, Moser B, et al. CCR5 is characteristic of Th1 lymphocytes. Nature. 1998;391:344–345.
    1. Muller KM, Jaunin F, Masouye I, Saurat JH, Hauser C. Th2 cells mediate IL-4-dependent local tissue inflammation. J Immunol. 1993;150:5576–5584.
    1. Kamogawa Y, Minasi LE, Carding SR, Bottomly K, Flavell RA. The relationship of IL-4- and IFNg-producing T cells studied by lineage ablation of IL-4-producing cells. Cell. 1993;75:985–995.
    1. Del Prete G. Human Th1 and Th2 lymphocytes: Their role in the pathophysiology of atopy. Allergy. 1992;47:450–455.
    1. D'Ambrosio D, Mariani M, Panina-Bordignon P, Sinigaglia F. Chemokines and their receptors guiding t lymphocyte recruitment in lung inflammation. Am J Respir Crit Care Med. 2001;164:1266–1275.
    1. Panina-Bordignon P, Papi A, Mariani M, Di Lucia P, Casoni G, et al. The C-C chemokine receptors CCR4 and CCR8 identify airway T cells of allergen-challenged atopic asthmatics. J Clin Invest. 2001;107:1357–1364.
    1. Cole KE, Strick CA, Paradis TJ, Ogborne KT, Loetscher M, et al. Interferon-inducible T cell alpha chemoattractant (I-TAC): A novel non-ELR CXC chemokine with potent activity on activated T cells through selective high affinity binding to CXCR3. J Exp Med. 1998;187:2009–2021.
    1. Loetscher P, Pellegrino A, Gong JH, Mattioli I, Loetscher M, et al. The ligands of CXC chemokine receptor 3, I-TAC, Mig, and IP10, are natural antagonists for CCR3. J Biol Chem. 2001;276:2986–2991.
    1. Zlotnik A, Yoshie O. Chemokines: A new classification system and their role in immunity. Immunity. 2000;12:121–127.
    1. Klier CM, Nelson EL, Cohen CD, Horuk R, Schlondorff D, et al. Chemokine-induced secretion of gelatinase B in primary human monocytes. Biol Chem. 2001;382:1405–1410.
    1. Pauwels RA, Buist AS, Calverley PM, Jenkins CR, Hurd SS. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. NHLBI/WHO Global Initiative for Chronic Obstructive Lung Disease (GOLD) Workshop summary. Am J Respir Crit Care Med. 2001;163:1256–1276.
    1. Uppaluri R, Mitsa T, Sonka M, Hoffman EA, McLennan G. Quantification of pulmonary emphysema from lung computed tomography images. Am J Respir Crit Care Med. 1997;156:248–254.
    1. Satoh K, Kobayashi T, Misao T, Hitani Y, Yamamoto Y, et al. CT assessment of subtypes of pulmonary emphysema in smokers. Chest. 2001;120:725–729.
    1. Madani A, Keyzer C, Gevenois PA. Quantitative computed tomography assessment of lung structure and function in pulmonary emphysema. Eur Respir J. 2001;18:720–730.
    1. Upham JW, McMenamin C, Schon HM, Robinson BW, Holt PG. Functional analysis of human bronchial mucosal T cells extracted with interleukin-2. Am J Respir Crit Care Med. 1994;149:1608–1613.
    1. Goodacre R, Davidson R, Singal D, Bienenstock J. Morphologic and functional characteristics of human intestinal lymphoid cells isolated by a mechanical technique. Gastroenterology. 1979;76:300–308.
    1. Fiocchi C. Lymphoid cells of the gastrointestinal tract. Isolation procedures. Acta Chir Scand Suppl. 1985;525:11–23.
    1. Corry DB, Rishi K, Kanellis J, Kiss A, Song Lz LZ, et al. Decreased allergic lung inflammatory cell egression and increased susceptibility to asphyxiation in MMP2-deficiency. Nat Immunol. 2002;3:347–353.
    1. Baggiolini M. Chemokines in pathology and medicine. J Intern Med. 2001;250:91–104.
    1. Renauld JC. New insights into the role of cytokines in asthma. J Clin Pathol. 2001;54:577–589.
    1. Padrid PA, Cozzi P, Leff AR. Cyclosporine A inhibits airway reactivity and remodeling after chronic antigen challenge in cats. Am J Respir Crit Care Med. 1996;154:1812–1818.
    1. Sternlicht MD, Werb Z. ECM Proteinases. In: Kreis T, Vale RD, editors. Guidebook to the extracellular matrix and adhesion proteins. Oxford: Oxford University Press; 1999. pp. 503–562.
    1. Wakugawa M, Nakamura K, Kakinuma T, Onai N, Matsushima K, et al. CC chemokine receptor 4 expression on peripheral blood CD4+ T cells reflects disease activity of atopic dermatitis. J Invest Dermatol. 2001;117:188–196.
    1. Saetta M, Mariani M, Panina-Bordignon P, Turato G, Buonsanti C, 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;165:1404–1409.
    1. Barnes PJ. Chronic obstructive pulmonary disease. N Engl J Med. 2000;343:269–280.
    1. Cawston T, Carrere S, Catterall J, Duggleby R, Elliott S, et al. Matrix metalloproteinases and TIMPs: Properties and implications for the treatment of chronic obstructive pulmonary disease. Novartis Found Symp. 2001;234:205–218.
    1. Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol. 2001;17:463–516.
    1. Kang T, Yi J, Guo A, Wang X, Overall CM, et al. Subcellular distribution and cytokine- and chemokine-regulated secretion of leukolysin/MT6-MMP/MMP-25 in neutrophils. J Biol Chem. 2001;276:21960–21968.
    1. Hautamaki RD, Kobayashi DK, Senior RM, Shapiro SD. Requirement for macrophage elastase for cigarette smoke-induced emphysema in mice. Science. 1997;277:2002–2004.
    1. Churg A, Zay K, Shay S, Xie C, Shapiro SD, et al. Acute cigarette smoke-induced connective tissue breakdown requires both neutrophils and macrophage metalloelastase in mice. Am J Respir Cell Mol Biol. 2002;27:368–374.
    1. Gronski TJ, Martin RL, Kobayashi DK, Walsh BC, Holman MC, et al. Hydrolysis of a broad spectrum of extracellular matrix proteins by human macrophage elastase. J Biol Chem. 1997;272:12189–12194.
    1. Liu Z, Zhou X, Shapiro SD, Shipley JM, Twining SS, et al. The serpin alpha1-proteinase inhibitor is a critical substrate for gelatinase B/MMP-9 in vivo. Cell. 2000;102:647–655.

Source: PubMed

3
S'abonner