Acute exacerbations of chronic obstructive pulmonary disease are associated with decreased CD4+ & CD8+ T cells and increased growth & differentiation factor-15 (GDF-15) in peripheral blood

Christine M Freeman, Carlos H Martinez, Jill C Todt, Fernando J Martinez, MeiLan K Han, Deborah L Thompson, Lisa McCloskey, Jeffrey L Curtis, Christine M Freeman, Carlos H Martinez, Jill C Todt, Fernando J Martinez, MeiLan K Han, Deborah L Thompson, Lisa McCloskey, Jeffrey L Curtis

Abstract

Background: Although T cells, especially CD8+, have been implicated in chronic obstructive pulmonary disease (COPD) pathogenesis, their role during acute exacerbations (AE-COPD) is uncertain.

Methods: We recruited subjects with COPD and a history of previous AE-COPD and studied them quarterly to collect blood and spontaneously expectorated sputum while stable. During exacerbations (defined by a change in symptoms plus physician diagnosis and altered medications), we collected blood and sputum before administering antibiotics or steroids. We used flow cytometry to identify leukocytes in peripheral blood, plus Luminex® analysis or ELISA to determine levels of inflammatory biomarkers in serum and sputum supernatants.

Results: Of 33 enrolled subjects, 13 participated in multiple stable visits and had ≥1 AE-COPD visit, yielding 18 events with paired data. Flow cytometric analyses of peripheral blood demonstrated decreased CD4+ and CD8+ T cells during AE-COPD (both absolute and as a percentage of all leukocytes) and significantly increased granulocytes, all of which correlated significantly with serum C-reactive protein (CRP) concentrations. No change was observed in other leukocyte populations during AE-COPD, although the percentage of BDCA-1+ dendritic cells expressing the activation markers CD40 and CD86 increased. During AE-COPD, sICAM-1, sVCAM-1, IL-10, IL-15 and GDF-15 increased in serum, while in sputum supernatants, CRP and TIMP-2 increased and TIMP-1 decreased.

Conclusions: The decrease in CD4+ and CD8+ T cells (but not other lymphocyte subsets) in peripheral blood during AE-COPD may indicate T cell extravasation into inflammatory sites or organized lymphoid tissues. GDF-15, a sensitive marker of cardiopulmonary stress that in other settings independently predicts reduced long-term survival, is acutely increased in AE-COPD. These results extend the concept that AE-COPD are systemic inflammatory events to which adaptive immune mechanisms contribute.

Trial registration: NCT00281216 , ClinicalTrials.gov.

Figures

Fig. 1
Fig. 1
AE-COPD confirmed by increases in BCSS total score and serum CRP levels. AE-COPD events were identified by physician diagnosis (after exclusion of pneumonia by chest radiograph) plus the clinical decision to prescribe oral steroids or antibiotics. Although these decisions were blinded to biomarker results, the diagnosis of AE-COPD was retrospectively confirmed by highly significant increases in (a) BCSS total scores and (b) serum CRP levels. Open circles represent individual subjects; mean ± SEM of the grouped data are shown by the single red circles. The Wilcoxon matched-pairs signed rank test was used to determine significant differences between visits
Fig. 2
Fig. 2
Peripheral blood CD4+ and CD8+ T cells decreased during AE-COPD while CD15+ granulocytes increased. Peripheral blood was collected during stable visits and during AE-COPD, prior to medication (n = 18 paired samples). Cells were stained and analyzed by flow cytometry. Results are shown for the individual cell type as a percentage of all CD45+ leukocytes (a, c, e) and as absolute cell numbers per μL (b, d, f). A, B. CD4+ T cells. C, D. CD8+ T cells. E, F. CD15+ cells; note difference in scale of panels E & F. Open circles represent individual subjects; mean ± SEM of the grouped data are shown by the single red circles. The Wilcoxon matched-pairs signed rank test was used to determine significant differences between visits
Fig. 3
Fig. 3
Representative graphs from individual subjects tracking percentages of CD4+ and CD8+ T cells during stable and exacerbation visits. Peripheral blood was collected during stable visits and during AE-COPD, prior to medication. Cells were stained and analyzed by flow cytometry. Panels a-e each depicted the data for a single subject; CD4+ T cells (upper panels, blue) and CD8+ T cells (lower panels, red) shown as a percentage of all CD45+ leukocytes. a, b. Two subjects with multiple AE-COPD; c, d. two subjects with a single AE-COPD; and e, subject with no AE-COPD. Stable visits are shown by open circles and AE-COPD are shown in solid circles labeled with “Ex”
Fig. 4
Fig. 4
CD40 and CD86 increased on BDCA-1+ DCs from peripheral blood during AE-COPD. Peripheral blood was collected during stable visits and during AE-COPD, prior to medication (n = 10 paired samples). Cells were stained and analyzed by flow cytometry. After gating on BDCA-1+ DCs, the percent of cells expressing (a) CD40 and (b) CD86 was determined. Open circles represent individual subjects; mean ± SEM of the grouped data are shown by the single red circles. The Wilcoxon matched-pairs signed rank test was used to determine significant differences between visits
Fig. 5
Fig. 5
Soluble adhesion molecules and inflammatory mediators increased while CCL11 decreased in the serum during AE-COPD. Serum was collected during stable visits and during AE-COPD, prior to medication (n = 18 paired samples). Samples were stored at −80 °C prior to analysis by Luminex® assay, except as noted. (a) sICAM-1, (b) sVCAM-1, (c) IL-10, (d) IL-15, (e) GDF-15 (by ELISA), (f) CCL11 are shown. Open circles represent individual subjects; mean ± SEM of the grouped data are shown by the single red circles. Note differences in units of measurement. The dotted line represents the minimal detectable concentration. The Wilcoxon matched-pairs signed rank test was used to determine significant differences between visits
Fig. 6
Fig. 6
CRP and TIMP-2 increased in sputum during AE-COPD, whereas TIMP-1 decreased. Sputum was collected during stable visits and during AE-COPD, prior to medication. After processing, sputum supernatants were stored at −80 °C prior to analysis by Luminex. (a) CRP, (b) TIMP-1, (c) TIMP-2 are shown (n = 10 paired samples for CRP and n = 7 paired samples for TIMP-1 and TIMP-2). Open circles represent individual subjects; mean ± SEM of the grouped data are shown by the single red circles. The dotted line represents the minimal detectable concentration. The Wilcoxon matched-pairs signed rank test was used to determine significant differences between visits

References

    1. Donaldson GC, Seemungal TA, Bhowmik A, Wedzicha JA. Relationship between exacerbation frequency and lung function decline in chronic obstructive pulmonary disease. Thorax. 2002;57:847–852. doi: 10.1136/thorax.57.10.847.
    1. Seemungal TA, Donaldson GC, Paul EA, Bestall JC, Jeffries DJ, Wedzicha JA. Effect of exacerbation on quality of life in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157:1418–1422. doi: 10.1164/ajrccm.157.5.9709032.
    1. Singanayagam A, Schembri S, Chalmers JD. Predictors of mortality in hospitalized adults with acute exacerbation of chronic obstructive pulmonary disease. Ann Am Thorac Soc. 2013;10:81–89. doi: 10.1513/AnnalsATS.201208-043OC.
    1. Soler-Cataluna JJ, Martinez-Garcia MA, Roman Sanchez P, Salcedo E, Navarro M, Ochando R. Severe acute exacerbations and mortality in patients with chronic obstructive pulmonary disease. Thorax. 2005;60:925–931. doi: 10.1136/thx.2005.040527.
    1. Toy EL, Gallagher KF, Stanley EL, Swensen AR, Duh MS. The economic impact of exacerbations of chronic obstructive pulmonary disease and exacerbation definition: a review. COPD. 2010;7:214–228. doi: 10.3109/15412555.2010.481697.
    1. Seemungal T, Harper-Owen R, Bhowmik A, Moric I, Sanderson G, Message S, et al. Respiratory viruses, symptoms, and inflammatory markers in acute exacerbations and stable chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;164:1618–23.
    1. Patel IS, Seemungal TA, Wilks M, Lloyd-Owen SJ, Donaldson GC, Wedzicha JA. Relationship between bacterial colonisation and the frequency, character, and severity of COPD exacerbations. Thorax. 2002;57:759–764. doi: 10.1136/thorax.57.9.759.
    1. MacNee W, Donaldson K. Exacerbations of COPD: environmental mechanisms. Chest. 2000;117:390S–397S. doi: 10.1378/chest.117.5_suppl_2.390S.
    1. Bafadhel M, McKenna S, Terry S, Mistry V, Reid C, Haldar P, et al. Acute exacerbations of chronic obstructive pulmonary disease: identification of biologic clusters and their biomarkers. Am J Respir Crit Care Med. 2011;184:662–71.
    1. Curtis JL, Freeman CM, Hogg JC. The immunopathogenesis of chronic obstructive pulmonary disease: insights from recent research. Proc Am Thorac Soc. 2007;4:512–521. doi: 10.1513/pats.200701-002FM.
    1. Freeman CM, Han MK, Martinez FJ, Murray S, Liu LX, Chensue SW, 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;184:6504–13.
    1. Freeman CM, Martinez FJ, Han MK, Washko Jr GR, McCubbrey AL, Chensue SW, et al. Lung CD8+ T cells in COPD have increased expression of bacterial TLRs. Respir Res. 2013;14:13.
    1. Saetta M, Baraldo S, Corbino L, Turato G, Braccioni F, Rea F, et al. CD8 + ve cells in the lungs of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1999;160:711–7.
    1. Mercer PF, Shute JK, Bhowmik A, Donaldson GC, Wedzicha JA, Warner JA. MMP-9, TIMP-1 and inflammatory cells in sputum from COPD patients during exacerbation. Respir Res. 2005;6:151. doi: 10.1186/1465-9921-6-151.
    1. Fujimoto K, Yasuo M, Urushibata K, Hanaoka M, Koizumi T, Kubo K. Airway inflammation during stable and acutely exacerbated chronic obstructive pulmonary disease. Eur Respir J. 2005;25:640–646. doi: 10.1183/09031936.05.00047504.
    1. Saetta M, Di Stefano A, Maestrelli P, Turato G, Ruggieri MP, Roggeri A, et al. Airway eosinophilia in chronic bronchitis during exacerbations. Am J Respir Crit Care Med. 1994;150:1646–52.
    1. Tsoumakidou M, Tzanakis N, Chrysofakis G, Kyriakou D, Siafakas NM. Changes in sputum T-lymphocyte subpopulations at the onset of severe exacerbations of chronic obstructive pulmonary disease. Respir Med. 2005;99:572–579. doi: 10.1016/j.rmed.2004.10.005.
    1. Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med. 2007;176:532–55.
    1. McCubbrey AL, Sonstein J, Ames TM, Freeman CM, Curtis JL. Glucocorticoids relieve collectin-driven suppression of apoptotic cell uptake in murine alveolar macrophages through downregulation of SIRPalpha. J Immunol. 2012;189:112–119. doi: 10.4049/jimmunol.1200984.
    1. Freeman CM, Crudgington S, Stolberg VR, Brown JP, Sonstein J, Alexis NE, et al. Design of a multi-center immunophenotyping analysis of peripheral blood, sputum and bronchoalveolar lavage fluid in the Subpopulations and Intermediate Outcome Measures in COPD Study (SPIROMICS). J Transl Med. 2015;13:19.
    1. Leidy NK, Rennard SI, Schmier J, Jones MK, Goldman M. The breathlessness, cough, and sputum scale: the development of empirically based guidelines for interpretation. Chest. 2003;124:2182–2191. doi: 10.1378/chest.124.6.2182.
    1. Hurst JR, Donaldson GC, Perera WR, Wilkinson TM, Bilello JA, Hagan GW, et al. Use of plasma biomarkers at exacerbation of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2006;174:867–74.
    1. Mueller SN, Gebhardt T, Carbone FR, Heath WR. Memory T cell subsets, migration patterns, and tissue residence. Annu Rev Immunol. 2013;31:137–161. doi: 10.1146/annurev-immunol-032712-095954.
    1. Purwar R, Campbell J, Murphy G, Richards WG, Clark RA, Kupper TS. Resident memory T cells (T(RM)) are abundant in human lung: diversity, function, and antigen specificity. PLoS One. 2011;6 doi: 10.1371/journal.pone.0016245.
    1. Hoser G, Kawiak J, Domagala-Kulawik J, Kopinski P, Droszcz W. Flow cytometric evaluation of lymphocyte subpopulations in BALF of healthy smokers and nonsmokers. Folia Histochem Cytobiol. 1999;37:25–30.
    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;145:711–22.
    1. Lofdahl MJ, Roos-Engstrand E, Pourazar J, Bucht A, Dahlen B, Elmberger G, et al. Increased intraepithelial T-cells in stable COPD. Respir Med. 2008;102:1812–8.
    1. Freeman CM, McCubbrey AL, Crudgington S, Nelson J, Martinez FJ, Han MK, et al. Basal gene expression by lung CD4+ T cells in chronic obstructive pulmonary disease identifies independent molecular correlates of airflow obstruction and emphysema extent. PLoS One. 2014;9, e96421.
    1. Freeman CM, Martinez FJ, Han MK, Ames TM, Chensue SW, Todt JC, et al. Lung dendritic cell expression of maturation molecules increases with worsening chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009;180:1179–88.
    1. Kheradmand F, Shan M, Corry DB. Smoking gun: mature dendritic cells in human lung provide clues to chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009;180:1166–1167. doi: 10.1164/rccm.200909-1391ED.
    1. Shan M, Cheng HF, Song LZ, Roberts L, Green L, Hacken-Bitar J, et al. Lung myeloid dendritic cells coordinately induce TH1 and TH17 responses in human emphysema. Sci Transl Med. 2009;1:4ra10.
    1. Turato G, Zuin R, Saetta M. Pathogenesis and pathology of COPD. Respiration. 2001;68:117–128. doi: 10.1159/000050478.
    1. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, et al. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004;350:2645–53.
    1. Paats MS, Bergen IM, Hoogsteden HC, van der Eerden MM, Hendriks RW. Systemic CD4+ and CD8+ T-cell cytokine profiles correlate with GOLD stage in stable COPD. Eur Respir J. 2012;40:330–337. doi: 10.1183/09031936.00079611.
    1. Masopust D, Schenkel JM. The integration of T cell migration, differentiation and function. Nat Rev Immunol. 2013;13:309–320. doi: 10.1038/nri3442.
    1. van der Strate BW, Postma DS, Brandsma CA, Melgert BN, Luinge MA, Geerlings M, et al. Cigarette smoke-induced emphysema: A role for the B cell? Am J Respir Crit Care Med. 2006;173:751–8.
    1. Corrigan CJ, Hartnell A, Kay AB. T lymphocyte activation in acute severe asthma. Lancet. 1988;1:1129–1132. doi: 10.1016/S0140-6736(88)91951-4.
    1. Couper D, Lavange LM, Han M, Barr RG, Bleecker E, Hoffman EA, et al. Design of the Subpopulations and Intermediate Outcomes in COPD Study (SPIROMICS). Thorax. 2013;69:491–4.
    1. Saltini C, Hance AJ, Ferrans VJ, Basset F, Bitterman PB, Crystal RG. Accurate quantification of cells recovered by bronchoalveolar lavage. Am Rev Respir Dis. 1984;130:650–658.
    1. Curtis JL, Kaltreider HB. Characterization of bronchoalveolar lymphocytes during a specific antibody-forming cell response in the lungs of mice. Am Rev Respir Dis. 1989;139:393–400. doi: 10.1164/ajrccm/139.2.393.
    1. Bootcov MR, Bauskin AR, Valenzuela SM, Moore AG, Bansal M, He XY, et al. MIC-1, a novel macrophage inhibitory cytokine, is a divergent member of the TGF-beta superfamily. Proc Natl Acad Sci U S A. 1997;94:11514–9.
    1. Lawton LN, Bonaldo MF, Jelenc PC, Qiu L, Baumes SA, Marcelino RA, et al. Identification of a novel member of the TGF-beta superfamily highly expressed in human placenta. Gene. 1997;203:17–26.
    1. Hromas R, Hufford M, Sutton J, Xu D, Li Y, Lu L. PLAB, a novel placental bone morphogenetic protein. Biochim Biophys Acta. 1997;1354:40–44. doi: 10.1016/S0167-4781(97)00122-X.
    1. Yokoyama-Kobayashi M, Saeki M, Sekine S, Kato S. Human cDNA encoding a novel TGF-beta superfamily protein highly expressed in placenta. J Biochem. 1997;122:622–626. doi: 10.1093/oxfordjournals.jbchem.a021798.
    1. Böttner M, Laaff M, Schechinger B, Rappold G, Unsicker K, Suter-Crazzolara C. Characterization of the rat, mouse, and human genes of growth/differentiation factor-15/macrophage inhibiting cytokine-1 (GDF-15/MIC-1) Gene. 1999;237:105–111. doi: 10.1016/S0378-1119(99)00309-1.
    1. Tong S, Marjono B, Brown DA, Mulvey S, Breit SN, Manuelpillai U, et al. Serum concentrations of macrophage inhibitory cytokine 1 (MIC 1) as a predictor of miscarriage. Lancet. 2004;363:129–30.
    1. Buckhaults P, Rago C, St Croix B, Romans KE, Saha S, Zhang L, et al. Secreted and cell surface genes expressed in benign and malignant colorectal tumors. Cancer Res. 2001;61:6996–7001.
    1. Bonaca MP, Morrow DA, Braunwald E, Cannon CP, Jiang S, Breher S, et al. Growth differentiation factor-15 and risk of recurrent events in patients stabilized after acute coronary syndrome: observations from PROVE IT-TIMI 22. Arterioscler Thromb Vasc Biol. 2011;31:203–10.
    1. Khan SQ, Ng K, Dhillon O, Kelly D, Quinn P, Squire IB, et al. Growth differentiation factor-15 as a prognostic marker in patients with acute myocardial infarction. Eur Heart J. 2009;30:1057–65.
    1. Anand IS, Kempf T, Rector TS, Tapken H, Allhoff T, Jantzen F, et al. Serial measurement of growth-differentiation factor-15 in heart failure: relation to disease severity and prognosis in the Valsartan Heart Failure Trial. Circulation. 2010;122:1387–95.
    1. Lankeit M, Kempf T, Dellas C, Cuny M, Tapken H, Peter T, et al. Growth differentiation factor-15 for prognostic assessment of patients with acute pulmonary embolism. Am J Respir Crit Care Med. 2008;177:1018–25.
    1. Nickel N, Kempf T, Tapken H, Tongers J, Laenger F, Lehmann U, et al. Growth differentiation factor-15 in idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med. 2008;178:534–41.
    1. Meadows CA, Risbano MG, Zhang L, Geraci MW, Tuder RM, Collier DH, et al. Increased expression of growth differentiation factor-15 in systemic sclerosis-associated pulmonary arterial hypertension. Chest. 2011;139:994–1002.
    1. Nickel N, Jonigk D, Kempf T, Bockmeyer CL, Maegel L, Rische J, et al. GDF-15 is abundantly expressed in plexiform lesions in patients with pulmonary arterial hypertension and affects proliferation and apoptosis of pulmonary endothelial cells. Respir Res. 2011;12:62.
    1. Wiklund FE, Bennet AM, Magnusson PK, Eriksson UK, Lindmark F, Wu L, et al. Macrophage inhibitory cytokine-1 (MIC-1/GDF15): a new marker of all-cause mortality. Aging Cell. 2010;9:1057–64.
    1. Zdrenghea MT, Mallia P, Johnston SL. Immunological pathways in virus-induced COPD exacerbations: a role for IL-15. Eur J Clin Invest. 2012;42:1010–1015. doi: 10.1111/j.1365-2362.2012.02672.x.
    1. Pletz MW, Ioanas M, de Roux A, Burkhardt O, Lode H. Reduced spontaneous apoptosis in peripheral blood neutrophils during exacerbation of COPD. Eur Respir J. 2004;23:532–537. doi: 10.1183/09031936.04.00089004.
    1. Schmidt-Ioanas M, Pletz MW, de Roux A, Lode H. Apoptosis of peripheral blood neutrophils in COPD exacerbation does not correlate with serum cytokines. Respir Med. 2006;100:639–647. doi: 10.1016/j.rmed.2005.08.011.
    1. Ropcke S, Holz O, Lauer G, Muller M, Rittinghausen S, Ernst P, et al. Repeatability of and relationship between potential COPD biomarkers in bronchoalveolar lavage, bronchial biopsies, serum, and induced sputum. PLoS One. 2012;7, e46207.
    1. Wouters EF, Groenewegen KH, Dentener MA, Vernooy JH. Systemic inflammation in chronic obstructive pulmonary disease: the role of exacerbations. Proc Am Thorac Soc. 2007;4:626–634. doi: 10.1513/pats.200706-071TH.
    1. Dev D, Wallace E, Sankaran R, Cunniffe J, Govan JR, Wathen CG, et al. Value of C-reactive protein measurements in exacerbations of chronic obstructive pulmonary disease. Respir Med. 1998;92:664–7.
    1. Tang Y, Guan Y, Liu Y, Sun J, Xu L, Jiang Y. The role of the serum IL-33/sST2 axis and inflammatory cytokines in chronic obstructive pulmonary disease. J Interferon Cytokine Res. 2014;34:162–168. doi: 10.1089/jir.2013.0063.
    1. Bathoorn E, Liesker JJ, Postma DS, Koeter GH, van der Toorn M, van der Heide S, et al. Change in inflammation in out-patient COPD patients from stable phase to a subsequent exacerbation. Int J Chron Obstruct Pulmon Dis. 2009;4:101–9.
    1. Wedzicha JA, Seemungal TA, MacCallum PK, Paul EA, Donaldson GC, Bhowmik A, et al. Acute exacerbations of chronic obstructive pulmonary disease are accompanied by elevations of plasma fibrinogen and serum IL-6 levels. Thromb Haemost. 2000;84:210–5.
    1. Tufvesson E, Ekberg M, Bjermer L. Inflammatory biomarkers in sputum predict COPD exacerbations. Lung. 2013;191:413–416. doi: 10.1007/s00408-013-9473-5.
    1. Mocchegiani E, Giacconi R, Costarelli L. Metalloproteases/anti-metalloproteases imbalance in chronic obstructive pulmonary disease: genetic factors and treatment implications. Curr Opin Pulm Med. 2011;17(Suppl 1):S11–19. doi: 10.1097/01.mcp.0000410743.98087.12.
    1. Savarin C, Bergmann CC, Hinton DR, Stohlman SA. MMP-independent role of TIMP-1 at the blood brain barrier during viral encephalomyelitis. ASN Neuro. 2013;5 doi: 10.1042/AN20130033.
    1. Ashutosh. Chao C, Borgmann K, Brew K, Ghorpade A. Tissue inhibitor of metalloproteinases-1 protects human neurons from staurosporine and HIV-1-induced apoptosis: mechanisms and relevance to HIV-1-associated dementia. Cell Death Dis. 2012;3:e332. doi: 10.1038/cddis.2012.54.
    1. Albert RK, Connett J, Bailey WC, Casaburi R, Cooper Jr JA, Criner GJ, et al. Azithromycin for prevention of exacerbations of COPD. N Engl J Med. 2011;365:689–98.
    1. Mutlu LC, Altintas N, Aydin M, Tulubas F, Oran M, Kucukyalin V, Kaplan G, Gurel A. Growth Differentiation Factor-15 Is a novel biomarker predicting acute exacerbation of chronic obstructive pulmonary disease. Inflammation. 2015;445:155-60.

Source: PubMed

3
Abonner