Skip to main content
Top
Published in: Seminars in Immunopathology 4/2016

Open Access 01-07-2016 | Review

COPD immunopathology

Authors: Gaetano Caramori, Paolo Casolari, Adam Barczyk, Andrew L. Durham, Antonino Di Stefano, Ian Adcock

Published in: Seminars in Immunopathology | Issue 4/2016

Login to get access

Abstract

The immunopathology of chronic obstructive pulmonary disease (COPD) is based on the innate and adaptive inflammatory immune responses to the chronic inhalation of cigarette smoking. In the last quarter of the century, the analysis of specimens obtained from the lower airways of COPD patients compared with those from a control group of age-matched smokers with normal lung function has provided novel insights on the potential pathogenetic role of the different cells of the innate and acquired immune responses and their pro/anti-inflammatory mediators and intracellular signalling pathways, contributing to a better knowledge of the immunopathology of COPD both during its stable phase and during its exacerbations. This also has provided a scientific rationale for new drugs discovery and targeting to the lower airways. This review summarises and discusses the immunopathology of COPD patients, of different severity, compared with control smokers with normal lung function.
Literature
1.
go back to reference Global Initiative for Chronic Obstructive Lung Disease (2016). Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. www.goldcopd.org (Updated 2016) Global Initiative for Chronic Obstructive Lung Disease (2016). Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. www.​goldcopd.​org (Updated 2016)
2.
go back to reference Gnatiuc L, Caramori G (2014) COPD in nonsmokers: the biomass hypothesis—to be or not to be? Eur Respir J 44:8–10PubMedCrossRef Gnatiuc L, Caramori G (2014) COPD in nonsmokers: the biomass hypothesis—to be or not to be? Eur Respir J 44:8–10PubMedCrossRef
4.
go back to reference Silverman EK, Sandhaus RA (2009) Clinical practice. Alpha1-antitrypsin deficiency. N Engl J Med 360:2749–2757PubMedCrossRef Silverman EK, Sandhaus RA (2009) Clinical practice. Alpha1-antitrypsin deficiency. N Engl J Med 360:2749–2757PubMedCrossRef
5.
go back to reference Birring SS, Brightling CE, Bradding P, Entwisle JJ, Vara DD, Grigg J, Wardlaw AJ, Pavord ID (2002) Clinical, radiologic, and induced sputum features of chronic obstructive pulmonary disease in nonsmokers: a descriptive study. Am J Respir Crit Care Med 166:1078–1083PubMedCrossRef Birring SS, Brightling CE, Bradding P, Entwisle JJ, Vara DD, Grigg J, Wardlaw AJ, Pavord ID (2002) Clinical, radiologic, and induced sputum features of chronic obstructive pulmonary disease in nonsmokers: a descriptive study. Am J Respir Crit Care Med 166:1078–1083PubMedCrossRef
6.
go back to reference Rivera RM, Cosio MG, Ghezzo H, Salazar M, Pérez-Padilla R (2008) Comparison of lung morphology in COPD secondary to cigarette and biomass smoke. Int J Tuberc Lung Dis 12:972–977PubMed Rivera RM, Cosio MG, Ghezzo H, Salazar M, Pérez-Padilla R (2008) Comparison of lung morphology in COPD secondary to cigarette and biomass smoke. Int J Tuberc Lung Dis 12:972–977PubMed
7.
go back to reference Barnes PJ, Celli BR (2009) Systemic manifestations and comorbidities of COPD. Eur Respir J 33:1165–1185PubMedCrossRef Barnes PJ, Celli BR (2009) Systemic manifestations and comorbidities of COPD. Eur Respir J 33:1165–1185PubMedCrossRef
8.
go back to reference Hogg JC (2004) Pathophysiology of airflow limitation in chronic obstructive pulmonary disease. Lancet 364:709–721PubMedCrossRef Hogg JC (2004) Pathophysiology of airflow limitation in chronic obstructive pulmonary disease. Lancet 364:709–721PubMedCrossRef
9.
go back to reference Van Brabandt H, Cauberghs M, Verbeken E, Moerman P, Lauweryns JM, Van de Woestijne KP (1983) Partitioning of pulmonary impedance in excised human and canine lungs. J Appl Physiol 55:1733–1742PubMed Van Brabandt H, Cauberghs M, Verbeken E, Moerman P, Lauweryns JM, Van de Woestijne KP (1983) Partitioning of pulmonary impedance in excised human and canine lungs. J Appl Physiol 55:1733–1742PubMed
11.
go back to reference Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Paré PD (2004) The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med 350:2645–2653PubMedCrossRef Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Paré PD (2004) The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med 350:2645–2653PubMedCrossRef
12.
go back to reference Hasegawa M, Nasuhara Y, Onodera Y, Makita H, Nagai K, Fuke S, Ito Y, Betsuyaku T, Nishimura M (2006) Airflow limitation and airway dimensions in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 173:1309–1315PubMedCrossRef Hasegawa M, Nasuhara Y, Onodera Y, Makita H, Nagai K, Fuke S, Ito Y, Betsuyaku T, Nishimura M (2006) Airflow limitation and airway dimensions in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 173:1309–1315PubMedCrossRef
14.
go back to reference Scherrer M (1961) Asthma, chronic bronchitis, and pulmonary emphysema. Ciba Symp 9:227–237PubMed Scherrer M (1961) Asthma, chronic bronchitis, and pulmonary emphysema. Ciba Symp 9:227–237PubMed
15.
go back to reference Barnes PJ (2014) Cellular and molecular mechanisms of chronic obstructive pulmonary disease. Clin Chest Med 35:71–86PubMedCrossRef Barnes PJ (2014) Cellular and molecular mechanisms of chronic obstructive pulmonary disease. Clin Chest Med 35:71–86PubMedCrossRef
16.
go back to reference Postma DS, Reddel HK, ten Hacken NH, van den Berge M (2014) Asthma and chronic obstructive pulmonary disease: similarities and differences. Clin Chest Med 35:143–156PubMedCrossRef Postma DS, Reddel HK, ten Hacken NH, van den Berge M (2014) Asthma and chronic obstructive pulmonary disease: similarities and differences. Clin Chest Med 35:143–156PubMedCrossRef
17.
go back to reference Caramori G, Casolari P, Giuffrè S, Barczyk A, Adcock I, Papi A (2011) COPD pathology in the small airways. Panminerva Med 53:51–70PubMed Caramori G, Casolari P, Giuffrè S, Barczyk A, Adcock I, Papi A (2011) COPD pathology in the small airways. Panminerva Med 53:51–70PubMed
18.
go back to reference Hogg JC, Timens W (2009) The pathology of chronic obstructive pulmonary disease. Annu Rev Pathol 4:435–459PubMedCrossRef Hogg JC, Timens W (2009) The pathology of chronic obstructive pulmonary disease. Annu Rev Pathol 4:435–459PubMedCrossRef
19.
go back to reference D’Anna SE, Asnaghi R, Caramori G, Appendini L, Rizzo M, Cavallaro C, Marino G, Cappello F, Balbi B, Di Stefano A (2012) High-resolution computed tomography quantitation of emphysema is correlated with selected lung function values in stable COPD. Respiration 83:383–390PubMedCrossRef D’Anna SE, Asnaghi R, Caramori G, Appendini L, Rizzo M, Cavallaro C, Marino G, Cappello F, Balbi B, Di Stefano A (2012) High-resolution computed tomography quantitation of emphysema is correlated with selected lung function values in stable COPD. Respiration 83:383–390PubMedCrossRef
20.
go back to reference Galbán CJ, Han MK, Boes JL, Chughtai KA, Meyer CR, Johnson TD, Galbán S, Rehemtulla A, Kazerooni EA, Martinez FJ, Ross BD (2012) Computed tomography-based biomarker provides unique signature for diagnosis of COPD phenotypes and disease progression. Nat Med 18:1711–1715PubMedPubMedCentralCrossRef Galbán CJ, Han MK, Boes JL, Chughtai KA, Meyer CR, Johnson TD, Galbán S, Rehemtulla A, Kazerooni EA, Martinez FJ, Ross BD (2012) Computed tomography-based biomarker provides unique signature for diagnosis of COPD phenotypes and disease progression. Nat Med 18:1711–1715PubMedPubMedCentralCrossRef
21.
go back to reference McDonough JE, Yuan R, Suzuki M, Seyednejad N, Elliott WM, Sanchez PG, Wright AC, Gefter WB, Litzky L, Coxson HO, Paré PD, Sin DD, Pierce RA, Woods JC, McWilliams AM, Mayo JR, Lam SC, Cooper JD, Hogg JC (2011) Small-airway obstruction and emphysema in chronic obstructive pulmonary disease. N Engl J Med 365:1567–1575PubMedPubMedCentralCrossRef McDonough JE, Yuan R, Suzuki M, Seyednejad N, Elliott WM, Sanchez PG, Wright AC, Gefter WB, Litzky L, Coxson HO, Paré PD, Sin DD, Pierce RA, Woods JC, McWilliams AM, Mayo JR, Lam SC, Cooper JD, Hogg JC (2011) Small-airway obstruction and emphysema in chronic obstructive pulmonary disease. N Engl J Med 365:1567–1575PubMedPubMedCentralCrossRef
22.
go back to reference Di Stefano A, Caramori G, Barczyk A, Vicari C, Brun P, Zanini A, Cappello F, Garofano E, Padovani A, Contoli M, Casolari P, Durham AL, Chung KF, Barnes PJ, Papi A, Adcock I, Balbi B (2014) Innate immunity but not NLRP3 inflammasome activation correlates with severity of stable COPD. Thorax 69:516–524PubMedPubMedCentralCrossRef Di Stefano A, Caramori G, Barczyk A, Vicari C, Brun P, Zanini A, Cappello F, Garofano E, Padovani A, Contoli M, Casolari P, Durham AL, Chung KF, Barnes PJ, Papi A, Adcock I, Balbi B (2014) Innate immunity but not NLRP3 inflammasome activation correlates with severity of stable COPD. Thorax 69:516–524PubMedPubMedCentralCrossRef
23.
go back to reference Saetta M, Turato G, Baraldo S, Zanin A, Braccioni F, Mapp CE, Maestrelli P, Cavallesco G, Papi A, Fabbri LM (2000) Goblet cell hyperplasia and epithelial inflammation in peripheral airways of smokers with both symptoms of chronic bronchitis and airflow limitation. Am J Respir Crit Care Med 161:1016–1021PubMedCrossRef Saetta M, Turato G, Baraldo S, Zanin A, Braccioni F, Mapp CE, Maestrelli P, Cavallesco G, Papi A, Fabbri LM (2000) Goblet cell hyperplasia and epithelial inflammation in peripheral airways of smokers with both symptoms of chronic bronchitis and airflow limitation. Am J Respir Crit Care Med 161:1016–1021PubMedCrossRef
25.
go back to reference Tzanakis N, Chrysofakis G, Tsoumakidou M, Kyriakou D, Tsiligianni J, Bouros D, Siafakas NM (2004) Induced sputum CD8+ T-lymphocyte subpopulations in chronic obstructive pulmonary disease. Respir Med 98:57–65PubMedCrossRef Tzanakis N, Chrysofakis G, Tsoumakidou M, Kyriakou D, Tsiligianni J, Bouros D, Siafakas NM (2004) Induced sputum CD8+ T-lymphocyte subpopulations in chronic obstructive pulmonary disease. Respir Med 98:57–65PubMedCrossRef
26.
go back to reference Chrysofakis G, Tzanakis N, Kyriakoy D, Tsoumakidou M, Tsiligianni I, Klimathianaki M, Siafakas NM (2004) Perforin expression and cytotoxic activity of sputum CD8+ lymphocytes in patients with COPD. Chest 125:71–76PubMedCrossRef Chrysofakis G, Tzanakis N, Kyriakoy D, Tsoumakidou M, Tsiligianni I, Klimathianaki M, Siafakas NM (2004) Perforin expression and cytotoxic activity of sputum CD8+ lymphocytes in patients with COPD. Chest 125:71–76PubMedCrossRef
27.
go back to reference Di Stefano A, Caramori G, Oates T, Capelli A, Lusuardi M, Gnemmi I, Ioli F, Chung KF, Donner CF, Barnes PJ, Adcock IM (2002) Increased expression of nuclear factor-kappaB in bronchial biopsies from smokers and patients with COPD. Eur Respir J 20:556–563PubMedCrossRef Di Stefano A, Caramori G, Oates T, Capelli A, Lusuardi M, Gnemmi I, Ioli F, Chung KF, Donner CF, Barnes PJ, Adcock IM (2002) Increased expression of nuclear factor-kappaB in bronchial biopsies from smokers and patients with COPD. Eur Respir J 20:556–563PubMedCrossRef
28.
go back to reference Di Stefano A, Capelli A, Lusuardi M, Caramori G, Balbo P, Ioli F, Sacco S, Gnemmi I, Brun P, Adcock IM, Balbi B, Barnes PJ, Chung KF, Donner CF (2001) Decreased T lymphocyte infiltration in bronchial biopsies of subjects with severe chronic obstructive pulmonary disease. Clin Exp Allergy 31:893–902PubMedCrossRef Di Stefano A, Capelli A, Lusuardi M, Caramori G, Balbo P, Ioli F, Sacco S, Gnemmi I, Brun P, Adcock IM, Balbi B, Barnes PJ, Chung KF, Donner CF (2001) Decreased T lymphocyte infiltration in bronchial biopsies of subjects with severe chronic obstructive pulmonary disease. Clin Exp Allergy 31:893–902PubMedCrossRef
29.
go back to reference Costabel U, Guzman J (1992) Effect of smoking on bronchoalveolar lavage constituents. Eur Respir J 5:776–779PubMed Costabel U, Guzman J (1992) Effect of smoking on bronchoalveolar lavage constituents. Eur Respir J 5:776–779PubMed
30.
go back to reference Siena L, Gjomarkaj M, Elliot J, Pace E, Bruno A, Baraldo S, Saetta M, Bonsignore MR, James A (2011) Reduced apoptosis of CD8+ T-lymphocytes in the airways of smokers with mild/moderate COPD. Respir Med 105:1491–1500PubMedCrossRef Siena L, Gjomarkaj M, Elliot J, Pace E, Bruno A, Baraldo S, Saetta M, Bonsignore MR, James A (2011) Reduced apoptosis of CD8+ T-lymphocytes in the airways of smokers with mild/moderate COPD. Respir Med 105:1491–1500PubMedCrossRef
31.
go back to reference Barceló B, Pons J, Ferrer JM, Sauleda J, Fuster A, Agustí AG (2008) Phenotypic characterisation of T-lymphocytes in COPD: abnormal CD4+CD25+ regulatory T-lymphocyte response to tobacco smoking. Eur Respir J 31:555–562PubMedCrossRef Barceló B, Pons J, Ferrer JM, Sauleda J, Fuster A, Agustí AG (2008) Phenotypic characterisation of T-lymphocytes in COPD: abnormal CD4+CD25+ regulatory T-lymphocyte response to tobacco smoking. Eur Respir J 31:555–562PubMedCrossRef
32.
go back to reference Cosio MG (2004) Autoimmunity, T-cells and STAT-4 in the pathogenesis of chronic obstructive pulmonary disease. Eur Respir J 24:3–5PubMedCrossRef Cosio MG (2004) Autoimmunity, T-cells and STAT-4 in the pathogenesis of chronic obstructive pulmonary disease. Eur Respir J 24:3–5PubMedCrossRef
33.
go back to reference Zhu J, Majumdar S, Qiu Y, Ansari T, Oliva A, Kips JC, Pauwels RA, De Rose V, Jeffery PK (2001) Interleukin-4 and interleukin-5 gene expression and inflammation in the mucus-secreting glands and subepithelial tissue of smokers with chronic bronchitis. Lack of relationship with CD8+ cells. Am J Respir Crit Care Med 164:2220–2228PubMedCrossRef Zhu J, Majumdar S, Qiu Y, Ansari T, Oliva A, Kips JC, Pauwels RA, De Rose V, Jeffery PK (2001) Interleukin-4 and interleukin-5 gene expression and inflammation in the mucus-secreting glands and subepithelial tissue of smokers with chronic bronchitis. Lack of relationship with CD8+ cells. Am J Respir Crit Care Med 164:2220–2228PubMedCrossRef
34.
go back to reference Halwani R, Al-Muhsen S, Hamid Q (2013) T helper 17 cells in airway diseases: from laboratory bench to bedside. Chest 143:494–501PubMedCrossRef Halwani R, Al-Muhsen S, Hamid Q (2013) T helper 17 cells in airway diseases: from laboratory bench to bedside. Chest 143:494–501PubMedCrossRef
35.
go back to reference Brusselle G, Bracke K (2014) Targeting immune pathways for therapy in asthma and chronic obstructive pulmonary disease. Ann Am Thorac Soc 11(Suppl 5):S322–S328PubMedCrossRef Brusselle G, Bracke K (2014) Targeting immune pathways for therapy in asthma and chronic obstructive pulmonary disease. Ann Am Thorac Soc 11(Suppl 5):S322–S328PubMedCrossRef
36.
go back to reference Vanaudenaerde BM, Verleden SE, Vos R, De Vleeschauwer SI, Willems-Widyastuti A, Geenens R, Van Raemdonck DE, Dupont LJ, Verbeken EK, Meyts I (2011) Innate and adaptive interleukin-17-producing lymphocytes in chronic inflammatory lung disorders. Am J Respir Crit Care Med 183:977–986PubMedCrossRef Vanaudenaerde BM, Verleden SE, Vos R, De Vleeschauwer SI, Willems-Widyastuti A, Geenens R, Van Raemdonck DE, Dupont LJ, Verbeken EK, Meyts I (2011) Innate and adaptive interleukin-17-producing lymphocytes in chronic inflammatory lung disorders. Am J Respir Crit Care Med 183:977–986PubMedCrossRef
37.
go back to reference Maneechotesuwan K, Kasetsinsombat K, Wongkajornsilp A, Barnes PJ (2013) Decreased indoleamine 2,3-dioxygenase activity and IL-10/IL-17A ratio in patients with COPD. Thorax 68:330–337PubMedCrossRef Maneechotesuwan K, Kasetsinsombat K, Wongkajornsilp A, Barnes PJ (2013) Decreased indoleamine 2,3-dioxygenase activity and IL-10/IL-17A ratio in patients with COPD. Thorax 68:330–337PubMedCrossRef
38.
go back to reference Chang Y, Al-Alwan L, Alshakfa S, Audusseau S, Mogas AK, Chouiali F, Nair P, Baglole CJ, Hamid Q, Eidelman DH (2014) Upregulation of IL-17A/F from human lung tissue explants with cigarette smoke exposure: implications for COPD. Respir Res 15:145PubMedPubMedCentralCrossRef Chang Y, Al-Alwan L, Alshakfa S, Audusseau S, Mogas AK, Chouiali F, Nair P, Baglole CJ, Hamid Q, Eidelman DH (2014) Upregulation of IL-17A/F from human lung tissue explants with cigarette smoke exposure: implications for COPD. Respir Res 15:145PubMedPubMedCentralCrossRef
39.
go back to reference Di Stefano A, Caramori G, Gnemmi I, Contoli M, Vicari C, Capelli A, Magno F, D’Anna SE, Zanini A, Brun P, Casolari P, Chung KF, Barnes PJ, Papi A, Adcock I, Balbi B (2009) T helper type 17-related cytokine expression is increased in the bronchial mucosa of stable chronic obstructive pulmonary disease patients. Clin Exp Immunol 157:316–324PubMedPubMedCentralCrossRef Di Stefano A, Caramori G, Gnemmi I, Contoli M, Vicari C, Capelli A, Magno F, D’Anna SE, Zanini A, Brun P, Casolari P, Chung KF, Barnes PJ, Papi A, Adcock I, Balbi B (2009) T helper type 17-related cytokine expression is increased in the bronchial mucosa of stable chronic obstructive pulmonary disease patients. Clin Exp Immunol 157:316–324PubMedPubMedCentralCrossRef
40.
go back to reference Eustace A, Smyth LJ, Mitchell L, Williamson K, Plumb J, Singh D (2011) Identification of cells expressing IL-17A and IL-17F in the lungs of patients with COPD. Chest 139:1089–1100PubMedCrossRef Eustace A, Smyth LJ, Mitchell L, Williamson K, Plumb J, Singh D (2011) Identification of cells expressing IL-17A and IL-17F in the lungs of patients with COPD. Chest 139:1089–1100PubMedCrossRef
41.
go back to reference Roos AB, Sandén C, Mori M, Bjermer L, Stampfli MR, Erjefält JS (2015) IL-17A is elevated in end-stage chronic obstructive pulmonary disease and contributes to cigarette smoke-induced lymphoid neogenesis. Am J Respir Crit Care Med 191:1232–1241PubMedCrossRef Roos AB, Sandén C, Mori M, Bjermer L, Stampfli MR, Erjefält JS (2015) IL-17A is elevated in end-stage chronic obstructive pulmonary disease and contributes to cigarette smoke-induced lymphoid neogenesis. Am J Respir Crit Care Med 191:1232–1241PubMedCrossRef
42.
go back to reference Zhang MQ, Wan Y, Jin Y, Xin JB, Zhang JC, Xiong XZ, Chen L, Chen G (2014) Cigarette smoking promotes inflammation in patients with COPD by affecting the polarization and survival of Th/Tregs through up-regulation of muscarinic receptor 3 and 5 expression. PLoS One 9, e112350PubMedPubMedCentralCrossRef Zhang MQ, Wan Y, Jin Y, Xin JB, Zhang JC, Xiong XZ, Chen L, Chen G (2014) Cigarette smoking promotes inflammation in patients with COPD by affecting the polarization and survival of Th/Tregs through up-regulation of muscarinic receptor 3 and 5 expression. PLoS One 9, e112350PubMedPubMedCentralCrossRef
43.
go back to reference Urboniene D, Babusyte A, Lötvall J, Sakalauskas R, Sitkauskiene B (2013) Distribution of γδ and other T-lymphocyte subsets in patients with chronic obstructive pulmonary disease and asthma. Respir Med 107:413–4723PubMedCrossRef Urboniene D, Babusyte A, Lötvall J, Sakalauskas R, Sitkauskiene B (2013) Distribution of γδ and other T-lymphocyte subsets in patients with chronic obstructive pulmonary disease and asthma. Respir Med 107:413–4723PubMedCrossRef
44.
go back to reference Freeman CM, McCubbrey AL, Crudgington S, Nelson J, Martinez FJ, Han MK, Washko GR Jr, Chensue SW, Arenberg DA, Meldrum CA, McCloskey L, Curtis JL (2014) 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 9, e96421PubMedPubMedCentralCrossRef Freeman CM, McCubbrey AL, Crudgington S, Nelson J, Martinez FJ, Han MK, Washko GR Jr, Chensue SW, Arenberg DA, Meldrum CA, McCloskey L, Curtis JL (2014) 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 9, e96421PubMedPubMedCentralCrossRef
45.
go back to reference Cosio MG, Saetta M, Agusti A (2009) Immunologic aspects of chronic obstructive pulmonary disease. N Engl J Med 360:2445–2454PubMedCrossRef Cosio MG, Saetta M, Agusti A (2009) Immunologic aspects of chronic obstructive pulmonary disease. N Engl J Med 360:2445–2454PubMedCrossRef
46.
go back to reference Bhat TA, Panzica L, Kalathil SG, Thanavala Y (2015) Immune dysfunction in patients with chronic obstructive pulmonary disease. Ann Am Thorac Soc 12(Suppl 2):S169–S175PubMedPubMedCentral Bhat TA, Panzica L, Kalathil SG, Thanavala Y (2015) Immune dysfunction in patients with chronic obstructive pulmonary disease. Ann Am Thorac Soc 12(Suppl 2):S169–S175PubMedPubMedCentral
47.
go back to reference Isajevs S, Taivans I, Strazda G, Kopeika U, Bukovskis M, Gordjusina V, Kratovska A (2009) Decreased FOXP3 expression in small airways of smokers with COPD. Eur Respir J 33:61–67PubMedCrossRef Isajevs S, Taivans I, Strazda G, Kopeika U, Bukovskis M, Gordjusina V, Kratovska A (2009) Decreased FOXP3 expression in small airways of smokers with COPD. Eur Respir J 33:61–67PubMedCrossRef
48.
go back to reference Hou J, Sun Y, Hao Y, Zhuo J, Liu X, Bai P, Han J, Zheng X, Zeng H (2013) Imbalance between subpopulations of regulatory T cells in COPD. Thorax 68:1131–1139PubMedCrossRef Hou J, Sun Y, Hao Y, Zhuo J, Liu X, Bai P, Han J, Zheng X, Zeng H (2013) Imbalance between subpopulations of regulatory T cells in COPD. Thorax 68:1131–1139PubMedCrossRef
49.
go back to reference Kalathil SG, Lugade AA, Pradhan V, Miller A, Parameswaran GI, Sethi S, Thanavala Y (2014) T-regulatory cells and programmed death 1+ T cells contribute to effector T-cell dysfunction in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 190:40–50PubMedPubMedCentralCrossRef Kalathil SG, Lugade AA, Pradhan V, Miller A, Parameswaran GI, Sethi S, Thanavala Y (2014) T-regulatory cells and programmed death 1+ T cells contribute to effector T-cell dysfunction in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 190:40–50PubMedPubMedCentralCrossRef
50.
go back to reference Li H, Liu Q, Jiang Y, Zhang Y, Zhang Y, Xiao W (2015) Disruption of th17/treg balance in the sputum of patients with chronic obstructive pulmonary disease. Am J Med Sci 349:392–397PubMedCrossRef Li H, Liu Q, Jiang Y, Zhang Y, Zhang Y, Xiao W (2015) Disruption of th17/treg balance in the sputum of patients with chronic obstructive pulmonary disease. Am J Med Sci 349:392–397PubMedCrossRef
51.
go back to reference Cappello F, Caramori G, Campanella C, Vicari C, Gnemmi I, Zanini A, Spanevello A, Capelli A, La Rocca G, Anzalone R, Bucchieri F, D’Anna SE, Ricciardolo FL, Brun P, Balbi B, Carone M, Zummo G, Conway de Macario E, Macario AJ, Di Stefano A (2011) Convergent sets of data from in vivo and in vitro methods point to an active role of Hsp60 in chronic obstructive pulmonary disease pathogenesis. PLoS One 6, e28200PubMedPubMedCentralCrossRef Cappello F, Caramori G, Campanella C, Vicari C, Gnemmi I, Zanini A, Spanevello A, Capelli A, La Rocca G, Anzalone R, Bucchieri F, D’Anna SE, Ricciardolo FL, Brun P, Balbi B, Carone M, Zummo G, Conway de Macario E, Macario AJ, Di Stefano A (2011) Convergent sets of data from in vivo and in vitro methods point to an active role of Hsp60 in chronic obstructive pulmonary disease pathogenesis. PLoS One 6, e28200PubMedPubMedCentralCrossRef
52.
go back to reference Pridgeon C, Bugeon L, Donnelly L, Straschil U, Tudhope SJ, Fenwick P, Lamb JR, Barnes PJ, Dallman MJ (2011) Regulation of IL-17 in chronic inflammation in the human lung. Clin Sci (Lond) 120:515–524CrossRef Pridgeon C, Bugeon L, Donnelly L, Straschil U, Tudhope SJ, Fenwick P, Lamb JR, Barnes PJ, Dallman MJ (2011) Regulation of IL-17 in chronic inflammation in the human lung. Clin Sci (Lond) 120:515–524CrossRef
53.
go back to reference Lee SH, Goswami S, Grudo A, Song LZ, Bandi V, Goodnight-White S, Green L, Hacken-Bitar J, Huh J, Bakaeen F, Coxson HO, Cogswell S, Storness-Bliss C, Corry DB, Kheradmand F (2007) Antielastin autoimmunity in tobacco smoking-induced emphysema. Nat Med 13:567–569PubMedCrossRef Lee SH, Goswami S, Grudo A, Song LZ, Bandi V, Goodnight-White S, Green L, Hacken-Bitar J, Huh J, Bakaeen F, Coxson HO, Cogswell S, Storness-Bliss C, Corry DB, Kheradmand F (2007) Antielastin autoimmunity in tobacco smoking-induced emphysema. Nat Med 13:567–569PubMedCrossRef
54.
go back to reference Kirkham PA, Caramori G, Casolari P, Papi AA, Edwards M, Shamji B, Triantaphyllopoulos K, Hussain F, Pinart M, Khan Y, Heinemann L, Stevens L, Yeadon M, Barnes PJ, Chung KF, Adcock IM (2011) Oxidative stress-induced antibodies to carbonyl-modified protein correlate with severity of chronic obstructive pulmonary disease. Am J Respir Crit Care Med 184:796–802PubMedPubMedCentralCrossRef Kirkham PA, Caramori G, Casolari P, Papi AA, Edwards M, Shamji B, Triantaphyllopoulos K, Hussain F, Pinart M, Khan Y, Heinemann L, Stevens L, Yeadon M, Barnes PJ, Chung KF, Adcock IM (2011) Oxidative stress-induced antibodies to carbonyl-modified protein correlate with severity of chronic obstructive pulmonary disease. Am J Respir Crit Care Med 184:796–802PubMedPubMedCentralCrossRef
55.
go back to reference Olloquequi J, Ferrer J, Montes JF, Rodríguez E, Montero MA, García-Valero J (2010) Differential lymphocyte infiltration in small airways and lung parenchyma in COPD patients. Respir Med 104:1310–1318PubMedCrossRef Olloquequi J, Ferrer J, Montes JF, Rodríguez E, Montero MA, García-Valero J (2010) Differential lymphocyte infiltration in small airways and lung parenchyma in COPD patients. Respir Med 104:1310–1318PubMedCrossRef
56.
go back to reference Brusselle GG, Demoor T, Bracke KR, Brandsma CA, Timens W (2009) Lymphoid follicles in (very) severe COPD: beneficial or harmful? Eur Respir J 34:219–230PubMedCrossRef Brusselle GG, Demoor T, Bracke KR, Brandsma CA, Timens W (2009) Lymphoid follicles in (very) severe COPD: beneficial or harmful? Eur Respir J 34:219–230PubMedCrossRef
57.
go back to reference van der Strate BW, Postma DS, Brandsma CA, Melgert BN, Luinge MA, Geerlings M, Hylkema MN, van den Berg A, Timens W, Kerstjens HA (2006) Cigarette smoke-induced emphysema: a role for the B cell? Am J Respir Crit Care Med 173:751–758PubMedCrossRef van der Strate BW, Postma DS, Brandsma CA, Melgert BN, Luinge MA, Geerlings M, Hylkema MN, van den Berg A, Timens W, Kerstjens HA (2006) Cigarette smoke-induced emphysema: a role for the B cell? Am J Respir Crit Care Med 173:751–758PubMedCrossRef
58.
go back to reference Polverino F, Baraldo S, Bazzan E, Agostini S, Turato G, Lunardi F, Balestro E, Damin M, Papi A, Maestrelli P, Calabrese F, Saetta M (2010) A novel insight into adaptive immunity in COPD: B cell activating factor belonging to the TNF family (BAFF). Am J Respir Crit Care Med 182:1011–1019PubMedCrossRef Polverino F, Baraldo S, Bazzan E, Agostini S, Turato G, Lunardi F, Balestro E, Damin M, Papi A, Maestrelli P, Calabrese F, Saetta M (2010) A novel insight into adaptive immunity in COPD: B cell activating factor belonging to the TNF family (BAFF). Am J Respir Crit Care Med 182:1011–1019PubMedCrossRef
59.
go back to reference Polverino F, Cosio BG, Pons J, Laucho-Contreras M, Tejera P, Iglesias A, Rios A, Jahn A, Sauleda J, Divo M, Pinto-Plata V, Sholl L, Rosas IO, Agustí A, Celli BR, Owen CA (2015) B cell-activating factor. An orchestrator of lymphoid follicles in severe chronic obstructive pulmonary disease. Am J Respir Crit Care Med 192:695–705PubMedPubMedCentralCrossRef Polverino F, Cosio BG, Pons J, Laucho-Contreras M, Tejera P, Iglesias A, Rios A, Jahn A, Sauleda J, Divo M, Pinto-Plata V, Sholl L, Rosas IO, Agustí A, Celli BR, Owen CA (2015) B cell-activating factor. An orchestrator of lymphoid follicles in severe chronic obstructive pulmonary disease. Am J Respir Crit Care Med 192:695–705PubMedPubMedCentralCrossRef
60.
go back to reference Seys LJ, Verhamme FM, Schinwald A, Hammad H, Cunoosamy DM, Bantsimba-Malanda C, Sabirsh A, McCall E, Flavell L, Herbst R, Provoost S, Lambrecht BN, Joos GF, Brusselle GG, Bracke KR (2015) Role of B cell-activating factor in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 192:706–718PubMedCrossRef Seys LJ, Verhamme FM, Schinwald A, Hammad H, Cunoosamy DM, Bantsimba-Malanda C, Sabirsh A, McCall E, Flavell L, Herbst R, Provoost S, Lambrecht BN, Joos GF, Brusselle GG, Bracke KR (2015) Role of B cell-activating factor in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 192:706–718PubMedCrossRef
61.
go back to reference Faner R, Cruz T, Casserras T, López-Giraldo A, Noell G, Coca I, Tal-Singer R, Miller B, Rodriguez-Roisin R, Spira A, Kalko SG, Agusti A (2016) Network analysis of lung transcriptomics reveals a distinct B cell signature in emphysema. Am J Respir Crit Care Med Faner R, Cruz T, Casserras T, López-Giraldo A, Noell G, Coca I, Tal-Singer R, Miller B, Rodriguez-Roisin R, Spira A, Kalko SG, Agusti A (2016) Network analysis of lung transcriptomics reveals a distinct B cell signature in emphysema. Am J Respir Crit Care Med
62.
go back to reference Hogg JC, Chu FSF, Tan WC, Sin DD, Patel SA, Pare PD, Martinez FJ, Rogers RM, Make BJ, Criner GJ, Cherniack RM, Sharafkhaneh A, Luketich JD, Coxson HO, Elliott WM, Sciurba FC (2007) Survival after lung volume reduction in chronic obstructive pulmonary disease: insights from small airway pathology. Am J Respir Crit Care Med 176:454–459PubMedPubMedCentralCrossRef Hogg JC, Chu FSF, Tan WC, Sin DD, Patel SA, Pare PD, Martinez FJ, Rogers RM, Make BJ, Criner GJ, Cherniack RM, Sharafkhaneh A, Luketich JD, Coxson HO, Elliott WM, Sciurba FC (2007) Survival after lung volume reduction in chronic obstructive pulmonary disease: insights from small airway pathology. Am J Respir Crit Care Med 176:454–459PubMedPubMedCentralCrossRef
63.
go back to reference Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW, Yates JC, Vestbo J, TORCH investigators (2007) Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl J Med 356:775–789PubMedCrossRef Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW, Yates JC, Vestbo J, TORCH investigators (2007) Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl J Med 356:775–789PubMedCrossRef
64.
go back to reference Focosi D, Bestagno M, Burrone O, Petrini M (2010) CD57+ T lymphocytes and functional immune deficiency. J Leukoc Biol 87:107–116PubMedCrossRef Focosi D, Bestagno M, Burrone O, Petrini M (2010) CD57+ T lymphocytes and functional immune deficiency. J Leukoc Biol 87:107–116PubMedCrossRef
65.
go back to reference Birch J, Anderson RK, Correia-Melo C, Jurk D, Hewitt G, Marques FM, Green NJ, Moisey E, Birrell MA, Belvisi MG, Black F, Taylor JJ, Fisher AJ, De Soyza A, Passos JF (2015) DNA damage response at telomeres contributes to lung aging and chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 309:L1124–L1137PubMedPubMedCentral Birch J, Anderson RK, Correia-Melo C, Jurk D, Hewitt G, Marques FM, Green NJ, Moisey E, Birrell MA, Belvisi MG, Black F, Taylor JJ, Fisher AJ, De Soyza A, Passos JF (2015) DNA damage response at telomeres contributes to lung aging and chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 309:L1124–L1137PubMedPubMedCentral
66.
go back to reference Olloquequi J, Montes JF, Prats A, Rodríguez E, Montero MA, García-Valero J, Ferrer J (2011) Significant increase of CD57+ cells in pulmonary lymphoid follicles of COPD patients. Eur Respir J 37:289–298PubMedCrossRef Olloquequi J, Montes JF, Prats A, Rodríguez E, Montero MA, García-Valero J, Ferrer J (2011) Significant increase of CD57+ cells in pulmonary lymphoid follicles of COPD patients. Eur Respir J 37:289–298PubMedCrossRef
67.
go back to reference Hiemstra PS (2013) Altered macrophage function in chronic obstructive pulmonary disease. Ann Am Thorac Soc 10(Suppl):S180–S185PubMedCrossRef Hiemstra PS (2013) Altered macrophage function in chronic obstructive pulmonary disease. Ann Am Thorac Soc 10(Suppl):S180–S185PubMedCrossRef
68.
go back to reference Chana KK, Fenwick PS, Nicholson AG, Barnes PJ, Donnelly LE (2014) Identification of a distinct glucocorticosteroid-insensitive pulmonary macrophage phenotype in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol 133:207–216PubMedCrossRef Chana KK, Fenwick PS, Nicholson AG, Barnes PJ, Donnelly LE (2014) Identification of a distinct glucocorticosteroid-insensitive pulmonary macrophage phenotype in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol 133:207–216PubMedCrossRef
69.
go back to reference Xue J, Schmidt SV, Sander J, Draffehn A, Krebs W, Quester I, De Nardo D, Gohel TD, Emde M, Schmidleithner L, Ganesan H, Nino-Castro A, Mallmann MR, Labzin L, Theis H, Kraut M, Beyer M, Latz E, Freeman TC, Ulas T, Schultze JL (2014) Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity 40:274–288PubMedPubMedCentralCrossRef Xue J, Schmidt SV, Sander J, Draffehn A, Krebs W, Quester I, De Nardo D, Gohel TD, Emde M, Schmidleithner L, Ganesan H, Nino-Castro A, Mallmann MR, Labzin L, Theis H, Kraut M, Beyer M, Latz E, Freeman TC, Ulas T, Schultze JL (2014) Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity 40:274–288PubMedPubMedCentralCrossRef
70.
go back to reference Kaku Y, Imaoka H, Morimatsu Y, Komohara Y, Ohnishi K, Oda H, Takenaka S, Matsuoka M, Kawayama T, Takeya M, Hoshino T (2014) Overexpression of CD163, CD204 and CD206 on alveolar macrophages in the lungs of patients with severe chronic obstructive pulmonary disease. PLoS One 9, e87400PubMedPubMedCentralCrossRef Kaku Y, Imaoka H, Morimatsu Y, Komohara Y, Ohnishi K, Oda H, Takenaka S, Matsuoka M, Kawayama T, Takeya M, Hoshino T (2014) Overexpression of CD163, CD204 and CD206 on alveolar macrophages in the lungs of patients with severe chronic obstructive pulmonary disease. PLoS One 9, e87400PubMedPubMedCentralCrossRef
71.
go back to reference Brusselle GG, Joos GF, Bracke KR (2011) New insights into the immunology of chronic obstructive pulmonary disease. Lancet 378:1015–1026PubMedCrossRef Brusselle GG, Joos GF, Bracke KR (2011) New insights into the immunology of chronic obstructive pulmonary disease. Lancet 378:1015–1026PubMedCrossRef
72.
go back to reference Tsoumakidou M, Bouloukaki I, Koutala H, Kouvidi K, Mitrouska I, Zakynthinos S, Tzanakis N, Jeffery PK, Siafakas NM (2009) Decreased sputum mature dendritic cells in healthy smokers and patients with chronic obstructive pulmonary disease. Int Arch Allergy Immunol 150:389–397PubMedCrossRef Tsoumakidou M, Bouloukaki I, Koutala H, Kouvidi K, Mitrouska I, Zakynthinos S, Tzanakis N, Jeffery PK, Siafakas NM (2009) Decreased sputum mature dendritic cells in healthy smokers and patients with chronic obstructive pulmonary disease. Int Arch Allergy Immunol 150:389–397PubMedCrossRef
73.
go back to reference Rogers AV, Adelroth E, Hattotuwa K, Dewar A, Jeffery PK (2008) Bronchial mucosal dendritic cells in smokers and ex-smokers with COPD: an electron microscopic study. Thorax 63:108–114PubMedCrossRef Rogers AV, Adelroth E, Hattotuwa K, Dewar A, Jeffery PK (2008) Bronchial mucosal dendritic cells in smokers and ex-smokers with COPD: an electron microscopic study. Thorax 63:108–114PubMedCrossRef
74.
go back to reference Zanini A, Spanevello A, Baraldo S, Majori M, Della Patrona S, Gumiero F, Aiello M, Olivieri D, Saetta M, Chetta A (2014) Decreased maturation of dendritic cells in the central airways of COPD patients is associated with VEGF, TGF-β and vascularity. Respiration 87:234–242PubMedCrossRef Zanini A, Spanevello A, Baraldo S, Majori M, Della Patrona S, Gumiero F, Aiello M, Olivieri D, Saetta M, Chetta A (2014) Decreased maturation of dendritic cells in the central airways of COPD patients is associated with VEGF, TGF-β and vascularity. Respiration 87:234–242PubMedCrossRef
75.
go back to reference Stoll P, Heinz AS, Bratke K, Bier A, Garbe K, Kuepper M, Virchow JC, Lommatzsch M (2014) Impact of smoking on dendritic cell phenotypes in the airway lumen of patients with COPD. Respir Res 15:48PubMedPubMedCentralCrossRef Stoll P, Heinz AS, Bratke K, Bier A, Garbe K, Kuepper M, Virchow JC, Lommatzsch M (2014) Impact of smoking on dendritic cell phenotypes in the airway lumen of patients with COPD. Respir Res 15:48PubMedPubMedCentralCrossRef
76.
go back to reference Tsoumakidou M, Koutsopoulos AV, Tzanakis N, Dambaki K, Tzortzaki E, Zakynthinos S, Jeffery PK, Siafakas NM (2009) Decreased small airway and alveolar CD83+ dendritic cells in COPD. Chest 136:726–733PubMedCrossRef Tsoumakidou M, Koutsopoulos AV, Tzanakis N, Dambaki K, Tzortzaki E, Zakynthinos S, Jeffery PK, Siafakas NM (2009) Decreased small airway and alveolar CD83+ dendritic cells in COPD. Chest 136:726–733PubMedCrossRef
77.
go back to reference Liao SX, Ding T, Rao XM, Sun DS, Sun PP, Wang YJ, Fu DD, Liu XL, Ou-Yang Y (2015) Cigarette smoke affects dendritic cell maturation in the small airways of patients with chronic obstructive pulmonary disease. Mol Med Rep 11:219–225PubMed Liao SX, Ding T, Rao XM, Sun DS, Sun PP, Wang YJ, Fu DD, Liu XL, Ou-Yang Y (2015) Cigarette smoke affects dendritic cell maturation in the small airways of patients with chronic obstructive pulmonary disease. Mol Med Rep 11:219–225PubMed
78.
go back to reference Demedts IK, Bracke KR, Van Pottelberge G, Testelmans D, Verleden GM, Vermassen FE, Joos GF, Brusselle GG (2007) Accumulation of dendritic cells and increased CCL20 levels in the airways of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 175:998–1005PubMedCrossRef Demedts IK, Bracke KR, Van Pottelberge G, Testelmans D, Verleden GM, Vermassen FE, Joos GF, Brusselle GG (2007) Accumulation of dendritic cells and increased CCL20 levels in the airways of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 175:998–1005PubMedCrossRef
79.
go back to reference Vassallo R, Walters PR, Lamont J, Kottom TJ, Yi ES, Limper AH (2010) Cigarette smoke promotes dendritic cell accumulation in COPD; a lung tissue research consortium study. Respir Res 11:45–57PubMedPubMedCentralCrossRef Vassallo R, Walters PR, Lamont J, Kottom TJ, Yi ES, Limper AH (2010) Cigarette smoke promotes dendritic cell accumulation in COPD; a lung tissue research consortium study. Respir Res 11:45–57PubMedPubMedCentralCrossRef
80.
go back to reference Van Pottelberge GR, Bracke KR, Demedts IK, De Rijck K, Reinartz SM, van Drunen CM, Verleden GM, Vermassen FE, Joos GF, Brusselle GG (2010) Selective accumulation of langerhans-type dendritic cells in small airways of patients with COPD. Respir Res 11:35PubMedPubMedCentralCrossRef Van Pottelberge GR, Bracke KR, Demedts IK, De Rijck K, Reinartz SM, van Drunen CM, Verleden GM, Vermassen FE, Joos GF, Brusselle GG (2010) Selective accumulation of langerhans-type dendritic cells in small airways of patients with COPD. Respir Res 11:35PubMedPubMedCentralCrossRef
81.
go back to reference Balzano G, Stefanelli F, Iorio C, De Felice A, Melillo EM, Martucci M, Melillo G (1999) Eosinophilic inflammation in stable chronic obstructive pulmonary disease. Relationship with neutrophils and airway function. Am J Respir Crit Care Med 160:1486–1492PubMedCrossRef Balzano G, Stefanelli F, Iorio C, De Felice A, Melillo EM, Martucci M, Melillo G (1999) Eosinophilic inflammation in stable chronic obstructive pulmonary disease. Relationship with neutrophils and airway function. Am J Respir Crit Care Med 160:1486–1492PubMedCrossRef
82.
go back to reference Thompson AB, Daughton D, Robbins RA, Ghafouri MA, Oehlerkin M, Rennard SI (1989) Intraluminal airway inflammation in chronic bronchitis: characterization and correlation with clinical parameters. Am Rev Respir Dis 140:1527–1537PubMedCrossRef Thompson AB, Daughton D, Robbins RA, Ghafouri MA, Oehlerkin M, Rennard SI (1989) Intraluminal airway inflammation in chronic bronchitis: characterization and correlation with clinical parameters. Am Rev Respir Dis 140:1527–1537PubMedCrossRef
83.
go back to reference Grashoff WF, Sont JK, Sterk PJ, Hiemstra PS, de Boer WI, Stolk J, Han J, van Krieken JM (1997) Chronic obstructive pulmonary disease: role of bronchial mast cells and macrophages. Am J Pathol 151:1785–1790PubMedPubMedCentral Grashoff WF, Sont JK, Sterk PJ, Hiemstra PS, de Boer WI, Stolk J, Han J, van Krieken JM (1997) Chronic obstructive pulmonary disease: role of bronchial mast cells and macrophages. Am J Pathol 151:1785–1790PubMedPubMedCentral
84.
go back to reference Baraldo S, Turato G, Badin C, Bazzan E, Beghé B, Zuin R, Calabrese F, Casoni G, Maestrelli P, Papi A, Fabbri LM, Saetta M (2004) Neutrophilic infiltration within the airway smooth muscle in patients with COPD. Thorax 59:308–312PubMedPubMedCentralCrossRef Baraldo S, Turato G, Badin C, Bazzan E, Beghé B, Zuin R, Calabrese F, Casoni G, Maestrelli P, Papi A, Fabbri LM, Saetta M (2004) Neutrophilic infiltration within the airway smooth muscle in patients with COPD. Thorax 59:308–312PubMedPubMedCentralCrossRef
85.
go back to reference Tsoumakidou M, Tsiligianni I, Tzanakis N (2011) Mechanisms of altered cell immunity and cytotoxicity in COPD. Curr Drug Targets 12:450–459PubMedCrossRef Tsoumakidou M, Tsiligianni I, Tzanakis N (2011) Mechanisms of altered cell immunity and cytotoxicity in COPD. Curr Drug Targets 12:450–459PubMedCrossRef
86.
go back to reference Upham JW, Stick SM (2006) Interactions between airway epithelial cells and dendritic cells: implications for the regulation of airway inflammation. Curr Drug Targets 7:541–545PubMedCrossRef Upham JW, Stick SM (2006) Interactions between airway epithelial cells and dendritic cells: implications for the regulation of airway inflammation. Curr Drug Targets 7:541–545PubMedCrossRef
87.
go back to reference Schulz C, Wolf K, Harth M, Kratzel K, Kunz-Schughart L, Pfeifer M (2003) Expression and release of interleukin-8 by human bronchial epithelial cells from patients with chronic obstructive pulmonary disease, smokers, and never-smokers. Respiration 70:254–261PubMedCrossRef Schulz C, Wolf K, Harth M, Kratzel K, Kunz-Schughart L, Pfeifer M (2003) Expression and release of interleukin-8 by human bronchial epithelial cells from patients with chronic obstructive pulmonary disease, smokers, and never-smokers. Respiration 70:254–261PubMedCrossRef
88.
go back to reference Suzuki T, Yamaya M, Sekizawa K, Hosoda M, Yamada N, Ishizuka S, Nakayama K, Yanai M, Numazaki Y, Sasaki H (2001) Bafilomycin A1 inhibits rhinovirus infection in human airway epithelium: effects on endosome and ICAM-1. Am J Physiol Lung Cell Mol Physiol 280:L1115–L1127PubMed Suzuki T, Yamaya M, Sekizawa K, Hosoda M, Yamada N, Ishizuka S, Nakayama K, Yanai M, Numazaki Y, Sasaki H (2001) Bafilomycin A1 inhibits rhinovirus infection in human airway epithelium: effects on endosome and ICAM-1. Am J Physiol Lung Cell Mol Physiol 280:L1115–L1127PubMed
90.
go back to reference de Marco R, Accordini S, Cerveri I, Corsico A, Antó JM, Künzli N, Janson C, Sunyer J, Jarvis D, Chinn S, Vermeire P, Svanes C, Ackermann-Liebrich U, Gislason T, Heinrich J, Leynaert B, Neukirch F, Schouten JP, Wjst M, Burney P (2007) Incidence of chronic obstructive pulmonary disease in a cohort of young adults according to the presence of chronic cough and phlegm. Am J Respir Crit Care Med 175:32–39PubMedCrossRef de Marco R, Accordini S, Cerveri I, Corsico A, Antó JM, Künzli N, Janson C, Sunyer J, Jarvis D, Chinn S, Vermeire P, Svanes C, Ackermann-Liebrich U, Gislason T, Heinrich J, Leynaert B, Neukirch F, Schouten JP, Wjst M, Burney P (2007) Incidence of chronic obstructive pulmonary disease in a cohort of young adults according to the presence of chronic cough and phlegm. Am J Respir Crit Care Med 175:32–39PubMedCrossRef
91.
go back to reference Caramori G, Casolari P, Di Gregorio C, Saetta M, Baraldo S, Boschetto P, Ito K, Fabbri LM, Barnes PJ, Adcock IM, Cavallesco G, Chung KF, Papi A (2009) MUC5AC expression is increased in bronchial submucosal glands of stable COPD patients. Histopathology 55:321–331PubMedCrossRef Caramori G, Casolari P, Di Gregorio C, Saetta M, Baraldo S, Boschetto P, Ito K, Fabbri LM, Barnes PJ, Adcock IM, Cavallesco G, Chung KF, Papi A (2009) MUC5AC expression is increased in bronchial submucosal glands of stable COPD patients. Histopathology 55:321–331PubMedCrossRef
92.
go back to reference Hovenberg HW, Davies JR, Carlstedt I (1996) Different mucins are produced by the surface epithelium and the submucosa in human trachea: identification of MUC5AC as a major mucin from the goblet cells. Biochem J 318(Pt 1):319–324PubMedPubMedCentralCrossRef Hovenberg HW, Davies JR, Carlstedt I (1996) Different mucins are produced by the surface epithelium and the submucosa in human trachea: identification of MUC5AC as a major mucin from the goblet cells. Biochem J 318(Pt 1):319–324PubMedPubMedCentralCrossRef
93.
go back to reference Schulz BL, Sloane AJ, Robinson LJ, Sebastian LT, Glanville AR, Song Y, Verkman AS, Harry JL, Packer NH, Karlsson NG (2005) Mucin glycosylation changes in cystic fibrosis lung disease are not manifest in submucosal gland secretions. Biochem J 387(Pt 3):911–919PubMedPubMedCentralCrossRef Schulz BL, Sloane AJ, Robinson LJ, Sebastian LT, Glanville AR, Song Y, Verkman AS, Harry JL, Packer NH, Karlsson NG (2005) Mucin glycosylation changes in cystic fibrosis lung disease are not manifest in submucosal gland secretions. Biochem J 387(Pt 3):911–919PubMedPubMedCentralCrossRef
94.
go back to reference Rose MC, Voynow JA (2006) Respiratory tract mucin genes and mucin glycoproteins in health and disease. Physiol Rev 86:245–278PubMedCrossRef Rose MC, Voynow JA (2006) Respiratory tract mucin genes and mucin glycoproteins in health and disease. Physiol Rev 86:245–278PubMedCrossRef
95.
go back to reference Hovenberg HW, Davies JR, Herrmann A, Lindén CJ, Carlstedt I (1996) MUC5AC, but not MUC2, is a prominent mucin in respiratory secretions. Glycoconj J 13:839–847PubMedCrossRef Hovenberg HW, Davies JR, Herrmann A, Lindén CJ, Carlstedt I (1996) MUC5AC, but not MUC2, is a prominent mucin in respiratory secretions. Glycoconj J 13:839–847PubMedCrossRef
96.
go back to reference Kirkham S, Sheehan JK, Knight D, Richardson PS, Thornton DJ (2002) Heterogeneity of airways mucus: variations in the amounts and glycoforms of the major oligomeric mucins MUC5AC and MUC5B. Biochem J 361(Pt 3):537–546PubMedPubMedCentralCrossRef Kirkham S, Sheehan JK, Knight D, Richardson PS, Thornton DJ (2002) Heterogeneity of airways mucus: variations in the amounts and glycoforms of the major oligomeric mucins MUC5AC and MUC5B. Biochem J 361(Pt 3):537–546PubMedPubMedCentralCrossRef
97.
go back to reference Kirkham S, Kolsum U, Rousseau K, Singh D, Vestbo J, Thornton DJ (2008) MUC5B is the major mucin in the gel phase of sputum in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 178:1033–1039PubMedPubMedCentralCrossRef Kirkham S, Kolsum U, Rousseau K, Singh D, Vestbo J, Thornton DJ (2008) MUC5B is the major mucin in the gel phase of sputum in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 178:1033–1039PubMedPubMedCentralCrossRef
98.
go back to reference Innes AL, Woodruff PG, Ferrando RE, Donnelly S, Dolganov GM, Lazarus SC, Fahy JV (2006) Epithelial mucin stores are increased in the large airways of smokers with airflow obstruction. Chest 130:1102–1108PubMedCrossRef Innes AL, Woodruff PG, Ferrando RE, Donnelly S, Dolganov GM, Lazarus SC, Fahy JV (2006) Epithelial mucin stores are increased in the large airways of smokers with airflow obstruction. Chest 130:1102–1108PubMedCrossRef
99.
go back to reference O’Donnell RA, Richter A, Ward J, Angco G, Mehta A, Rousseau K, Swallow DM, Holgate ST, Djukanovic R, Davies DE, Wilson SJ (2004) Expression of ErbB receptors and mucins in the airways of long term current smokers. Thorax 59:1032–1040PubMedPubMedCentralCrossRef O’Donnell RA, Richter A, Ward J, Angco G, Mehta A, Rousseau K, Swallow DM, Holgate ST, Djukanovic R, Davies DE, Wilson SJ (2004) Expression of ErbB receptors and mucins in the airways of long term current smokers. Thorax 59:1032–1040PubMedPubMedCentralCrossRef
100.
go back to reference Silvers GW, Maisel JC, Petty TL, Mitchell RS (1973) Reduction in peripheral airway resistance in excised emphysematous lungs. Chest 63(Suppl):32S–33SCrossRef Silvers GW, Maisel JC, Petty TL, Mitchell RS (1973) Reduction in peripheral airway resistance in excised emphysematous lungs. Chest 63(Suppl):32S–33SCrossRef
101.
go back to reference Caramori G, Di Gregorio C, Carlstedt I, Casolari P, Guzzinati I, Adcock IM, Barnes PJ, Ciaccia A, Cavallesco G, Chung KF, Papi A (2004) Mucin expression in peripheral airways of patients with chronic obstructive pulmonary disease. Histopathology 45:477–484PubMedCrossRef Caramori G, Di Gregorio C, Carlstedt I, Casolari P, Guzzinati I, Adcock IM, Barnes PJ, Ciaccia A, Cavallesco G, Chung KF, Papi A (2004) Mucin expression in peripheral airways of patients with chronic obstructive pulmonary disease. Histopathology 45:477–484PubMedCrossRef
102.
go back to reference Di Comite G, Grazia Sabbadini M, Corti A, Rovere-Querini P, Manfredi AA (2007) Conversation galante: how the immune and the neuroendocrine systems talk to each other. Autoimmun Rev 7:23–29PubMedCrossRef Di Comite G, Grazia Sabbadini M, Corti A, Rovere-Querini P, Manfredi AA (2007) Conversation galante: how the immune and the neuroendocrine systems talk to each other. Autoimmun Rev 7:23–29PubMedCrossRef
103.
go back to reference Profita M, Albano GD, Riccobono L, Di Sano C, Montalbano AM, Gagliardo R, Anzalone G, Bonanno A, Pieper MP, Gjomarkaj M (2014) Increased levels of Th17 cells are associated with non-neuronal acetylcholine in COPD patients. Immunobiology 219:392–401PubMedCrossRef Profita M, Albano GD, Riccobono L, Di Sano C, Montalbano AM, Gagliardo R, Anzalone G, Bonanno A, Pieper MP, Gjomarkaj M (2014) Increased levels of Th17 cells are associated with non-neuronal acetylcholine in COPD patients. Immunobiology 219:392–401PubMedCrossRef
104.
go back to reference Gu X, Karp PH, Brody SL, Pierce RA, Welsh MJ, Holtzman MJ, Ben-Shahar Y (2014) Chemosensory functions for pulmonary neuroendocrine cells. Am J Respir Cell Mol Biol 50:637–646PubMedPubMedCentralCrossRef Gu X, Karp PH, Brody SL, Pierce RA, Welsh MJ, Holtzman MJ, Ben-Shahar Y (2014) Chemosensory functions for pulmonary neuroendocrine cells. Am J Respir Cell Mol Biol 50:637–646PubMedPubMedCentralCrossRef
105.
go back to reference Vatrella A, Montagnani S, Calabrese C, Parrella R, Pelaia G, Biscione GL, Corcione N, Marsico SA, Guerra G (2010) Neuropeptide expression in the airways of COPD patients and smokers with normal lung function. J Biol Regul Homeost Agents 24:425–432PubMed Vatrella A, Montagnani S, Calabrese C, Parrella R, Pelaia G, Biscione GL, Corcione N, Marsico SA, Guerra G (2010) Neuropeptide expression in the airways of COPD patients and smokers with normal lung function. J Biol Regul Homeost Agents 24:425–432PubMed
106.
go back to reference Lucchini RE, Facchini F, Turato G, Saetta M, Caramori G, Ciaccia A, Maestrelli P, Springall DR, Polak JM, Fabbri L, Mapp CE (1997) Increased VIP-positive nerve fibers in the mucous glands of subjects with chronic bronchitis. Am J Respir Crit Care Med 156:1963–68PubMedCrossRef Lucchini RE, Facchini F, Turato G, Saetta M, Caramori G, Ciaccia A, Maestrelli P, Springall DR, Polak JM, Fabbri L, Mapp CE (1997) Increased VIP-positive nerve fibers in the mucous glands of subjects with chronic bronchitis. Am J Respir Crit Care Med 156:1963–68PubMedCrossRef
107.
go back to reference Miotto D, Boschetto P, Bononi I, Zeni E, Cavallesco G, Fabbri LM, Mapp CE (2004) Vasoactive intestinal peptide receptors in the airways of smokers with chronic bronchitis. Eur Respir J 24:958–963PubMedCrossRef Miotto D, Boschetto P, Bononi I, Zeni E, Cavallesco G, Fabbri LM, Mapp CE (2004) Vasoactive intestinal peptide receptors in the airways of smokers with chronic bronchitis. Eur Respir J 24:958–963PubMedCrossRef
108.
go back to reference Calabrese F, Giacometti C, Beghe B, Rea F, Loy M, Zuin R, Marulli G, Baraldo S, Saetta M, Valente M (2005) Marked alveolar apoptosis/proliferation imbalance in end-stage emphysema. Respir Res 6:14PubMedPubMedCentralCrossRef Calabrese F, Giacometti C, Beghe B, Rea F, Loy M, Zuin R, Marulli G, Baraldo S, Saetta M, Valente M (2005) Marked alveolar apoptosis/proliferation imbalance in end-stage emphysema. Respir Res 6:14PubMedPubMedCentralCrossRef
109.
go back to reference Kasahara Y, Tuder RM, Cool CD, Lynch DA, Flores SC, Voelkel NF (2001) Endothelial cell death and decreased expression of vascular endothelial growth factor and vascular endothelial growth factor receptor 2 in emphysema. Am J Respir Crit Care Med 163:737–744PubMedCrossRef Kasahara Y, Tuder RM, Cool CD, Lynch DA, Flores SC, Voelkel NF (2001) Endothelial cell death and decreased expression of vascular endothelial growth factor and vascular endothelial growth factor receptor 2 in emphysema. Am J Respir Crit Care Med 163:737–744PubMedCrossRef
110.
111.
go back to reference Suzuki M, Betsuyaku T, Nagai K, Fuke S, Nasuhara Y, Kaga K, Kondo S, Hamamura I, Hata J, Takahashi H, Nishimura M (2008) Decreased airway expression of vascular endothelial growth factor in cigarette smoke-induced emphysema in mice and COPD patients. Inhal Toxicol 20:349–359PubMedCrossRef Suzuki M, Betsuyaku T, Nagai K, Fuke S, Nasuhara Y, Kaga K, Kondo S, Hamamura I, Hata J, Takahashi H, Nishimura M (2008) Decreased airway expression of vascular endothelial growth factor in cigarette smoke-induced emphysema in mice and COPD patients. Inhal Toxicol 20:349–359PubMedCrossRef
112.
go back to reference Marwick JA, Stevenson CS, Giddings J, MacNee W, Butler K, Rahman I, Kirkham PA (2006) Cigarette smoke disrupts VEGF165-VEGFR-2 receptor signaling complex in rat lungs and patients with COPD: morphological impact of VEGFR-2 inhibition. Am J Physiol Lung Cell Mol Physiol 290:L897–L908PubMedCrossRef Marwick JA, Stevenson CS, Giddings J, MacNee W, Butler K, Rahman I, Kirkham PA (2006) Cigarette smoke disrupts VEGF165-VEGFR-2 receptor signaling complex in rat lungs and patients with COPD: morphological impact of VEGFR-2 inhibition. Am J Physiol Lung Cell Mol Physiol 290:L897–L908PubMedCrossRef
113.
go back to reference Santos S, Peinado VI, Ramirez J, Morales-Blanhir J, Bastos R, Roca J, Rodriguez-Roisin R, Barbera JA (2003) Enhanced expression of vascular endothelial growth factor in pulmonary arteries of smokers and patients with moderate chronic obstructive pulmonary disease. Am J Respir Crit Care Med 167:1250–1256PubMedCrossRef Santos S, Peinado VI, Ramirez J, Morales-Blanhir J, Bastos R, Roca J, Rodriguez-Roisin R, Barbera JA (2003) Enhanced expression of vascular endothelial growth factor in pulmonary arteries of smokers and patients with moderate chronic obstructive pulmonary disease. Am J Respir Crit Care Med 167:1250–1256PubMedCrossRef
114.
go back to reference Barberà JA, Blanco I (2009) Pulmonary hypertension in patients with chronic obstructive pulmonary disease: advances in pathophysiology and management. Drugs 69:1153–1171PubMedCrossRef Barberà JA, Blanco I (2009) Pulmonary hypertension in patients with chronic obstructive pulmonary disease: advances in pathophysiology and management. Drugs 69:1153–1171PubMedCrossRef
115.
go back to reference Zakynthinos E, Daniil Z, Papanikolaou J, Makris D (2011) Pulmonary hypertension in COPD: pathophysiology and therapeutic targets. Curr Drug Targets 12:501–513PubMedCrossRef Zakynthinos E, Daniil Z, Papanikolaou J, Makris D (2011) Pulmonary hypertension in COPD: pathophysiology and therapeutic targets. Curr Drug Targets 12:501–513PubMedCrossRef
116.
go back to reference Rowan SC, Keane MP, Gaine S, McLoughlin P (2016) Hypoxic pulmonary hypertension in chronic lung diseases: novel vasoconstrictor pathways. Lancet Respir Med 4(3):225–236PubMedCrossRef Rowan SC, Keane MP, Gaine S, McLoughlin P (2016) Hypoxic pulmonary hypertension in chronic lung diseases: novel vasoconstrictor pathways. Lancet Respir Med 4(3):225–236PubMedCrossRef
117.
go back to reference Kew KM, Seniukovich A (2014) Inhaled steroids and risk of pneumonia for chronic obstructive pulmonary disease. Cochrane Database Syst Rev 3, CD010115 Kew KM, Seniukovich A (2014) Inhaled steroids and risk of pneumonia for chronic obstructive pulmonary disease. Cochrane Database Syst Rev 3, CD010115
118.
go back to reference Liapikou A, Toumbis M, Torres A (2015) Managing the safety of inhaled corticosteroids in COPD and the risk of pneumonia. Expert Opin Drug Saf 14:1237–1247PubMedCrossRef Liapikou A, Toumbis M, Torres A (2015) Managing the safety of inhaled corticosteroids in COPD and the risk of pneumonia. Expert Opin Drug Saf 14:1237–1247PubMedCrossRef
119.
go back to reference Jen R, Rennard SI, Sin DD (2012) Effects of inhaled corticosteroids on airway inflammation in chronic obstructive pulmonary disease: a systematic review and meta-analysis. Int J Chron Obstruct Pulmon Dis 7:587–595PubMedPubMedCentral Jen R, Rennard SI, Sin DD (2012) Effects of inhaled corticosteroids on airway inflammation in chronic obstructive pulmonary disease: a systematic review and meta-analysis. Int J Chron Obstruct Pulmon Dis 7:587–595PubMedPubMedCentral
120.
go back to reference Ozol D, Aysan T, Solak ZA, Mogulkoc N, Veral A, Sebik F (2005) The effect of inhaled corticosteroids on bronchoalveolar lavage cells and IL-8 levels in stable COPD patients. Respir Med 99:1494–1500PubMedCrossRef Ozol D, Aysan T, Solak ZA, Mogulkoc N, Veral A, Sebik F (2005) The effect of inhaled corticosteroids on bronchoalveolar lavage cells and IL-8 levels in stable COPD patients. Respir Med 99:1494–1500PubMedCrossRef
121.
go back to reference Verhoeven GT, Garrelds IM, Hoogsteden HC, Zijlstra FJ (2001) Effects of fluticasone propionate inhalation on levels of arachidonic acid metabolites in patients with chronic obstructive pulmonary disease. Mediat Inflamm 10:21–26CrossRef Verhoeven GT, Garrelds IM, Hoogsteden HC, Zijlstra FJ (2001) Effects of fluticasone propionate inhalation on levels of arachidonic acid metabolites in patients with chronic obstructive pulmonary disease. Mediat Inflamm 10:21–26CrossRef
122.
go back to reference Keatings VM, Barnes PJ (1997) Granulocyte activation markers in induced sputum: comparison between chronic obstructive pulmonary disease, asthma, and normal subjects. Am J Respir Crit Care Med 155:449–453PubMedCrossRef Keatings VM, Barnes PJ (1997) Granulocyte activation markers in induced sputum: comparison between chronic obstructive pulmonary disease, asthma, and normal subjects. Am J Respir Crit Care Med 155:449–453PubMedCrossRef
123.
go back to reference Barnes PJ, Chowdhury B, Kharitonov SA, Magnussen H, Page CP, Postma D, Saeatta M (2006) Pulmonary biomarkers in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 174:6–14PubMedCrossRef Barnes PJ, Chowdhury B, Kharitonov SA, Magnussen H, Page CP, Postma D, Saeatta M (2006) Pulmonary biomarkers in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 174:6–14PubMedCrossRef
124.
go back to reference Bourbeau J, Christodoulopoulos P, Maltais F, Yamauchi Y, Olivenstein R, Hamid Q (2007) Effect of salmeterol/fluticasone propionate on airway inflammation in COPD: a randomised controlled trial. Thorax 62:938–943PubMedPubMedCentralCrossRef Bourbeau J, Christodoulopoulos P, Maltais F, Yamauchi Y, Olivenstein R, Hamid Q (2007) Effect of salmeterol/fluticasone propionate on airway inflammation in COPD: a randomised controlled trial. Thorax 62:938–943PubMedPubMedCentralCrossRef
125.
go back to reference Lapperre TS, Snoeck-Stroband JB, Gosman MM, Jansen DF, van Schadewijk A, Thiadens HA, Vonk JM, Boezen HM, Ten Hacken NH, Sont JK, Rabe KF, Kerstjens HA, Hiemstra PS, Timens W, Postma DS, Sterk PJ (2009) Effect of fluticasone with and without salmeterol on pulmonary outcomes in chronic obstructive pulmonary disease: a randomized trial. Ann Intern Med 151:517–527PubMedCrossRef Lapperre TS, Snoeck-Stroband JB, Gosman MM, Jansen DF, van Schadewijk A, Thiadens HA, Vonk JM, Boezen HM, Ten Hacken NH, Sont JK, Rabe KF, Kerstjens HA, Hiemstra PS, Timens W, Postma DS, Sterk PJ (2009) Effect of fluticasone with and without salmeterol on pulmonary outcomes in chronic obstructive pulmonary disease: a randomized trial. Ann Intern Med 151:517–527PubMedCrossRef
126.
go back to reference Lommatzsch M, Kraeft U, Troebs L, Garbe K, Bier A, Stoll P, Klammt S, Kuepper M, Bratke K, Virchow JC (2013) Fluticasone impact on airway dendritic cells in smokers: a randomized controlled trial. Respir Res 14:114PubMedPubMedCentralCrossRef Lommatzsch M, Kraeft U, Troebs L, Garbe K, Bier A, Stoll P, Klammt S, Kuepper M, Bratke K, Virchow JC (2013) Fluticasone impact on airway dendritic cells in smokers: a randomized controlled trial. Respir Res 14:114PubMedPubMedCentralCrossRef
129.
go back to reference Caramori G, Di Stefano A, Casolari P, Kirkham PA, Padovani A, Chung KF, Papi A, Adcock IM (2013) Chemokines and chemokine receptors blockers as new drugs for the treatment of chronic obstructive pulmonary disease. Curr Med Chem 20:4317–4349PubMedCrossRef Caramori G, Di Stefano A, Casolari P, Kirkham PA, Padovani A, Chung KF, Papi A, Adcock IM (2013) Chemokines and chemokine receptors blockers as new drugs for the treatment of chronic obstructive pulmonary disease. Curr Med Chem 20:4317–4349PubMedCrossRef
130.
go back to reference Caramori G, Adcock IM, Di Stefano A, Chung KF (2014) Cytokine inhibition in the treatment of COPD. Int J Chron Obstruct Pulmon Dis 9:397–412PubMedPubMedCentral Caramori G, Adcock IM, Di Stefano A, Chung KF (2014) Cytokine inhibition in the treatment of COPD. Int J Chron Obstruct Pulmon Dis 9:397–412PubMedPubMedCentral
132.
go back to reference Papi A, Luppi F, Franco F, Fabbri LM (2006) Pathophysiology of exacerbations of chronic obstructive pulmonary disease. Proc Am Thorac Soc 3:245–251PubMedCrossRef Papi A, Luppi F, Franco F, Fabbri LM (2006) Pathophysiology of exacerbations of chronic obstructive pulmonary disease. Proc Am Thorac Soc 3:245–251PubMedCrossRef
133.
135.
go back to reference Ricciardolo FL, Caramori G, Ito K, Capelli A, Brun P, Abatangelo G, Papi A, Chung KF, Adcock I, Barnes PJ, Donner CF, Rossi A, Di Stefano A (2005) Nitrosative stress in the bronchial mucosa of severe chronic obstructive pulmonary disease. J Allergy Clin Immunol 116:1028–1035PubMedCrossRef Ricciardolo FL, Caramori G, Ito K, Capelli A, Brun P, Abatangelo G, Papi A, Chung KF, Adcock I, Barnes PJ, Donner CF, Rossi A, Di Stefano A (2005) Nitrosative stress in the bronchial mucosa of severe chronic obstructive pulmonary disease. J Allergy Clin Immunol 116:1028–1035PubMedCrossRef
136.
go back to reference Wiegman CH, Michaeloudes C, Haji G, Narang P, Clarke CJ, Russell KE, Bao W, Pavlidis S, Barnes PJ, Kanerva J, Bittner A, Rao N, Murphy MP, Kirkham PA, Chung KF, Adcock IM, COPDMAP (2015) Oxidative stress-induced mitochondrial dysfunction drives inflammation and airway smooth muscle remodeling in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol 136:769–780PubMedPubMedCentralCrossRef Wiegman CH, Michaeloudes C, Haji G, Narang P, Clarke CJ, Russell KE, Bao W, Pavlidis S, Barnes PJ, Kanerva J, Bittner A, Rao N, Murphy MP, Kirkham PA, Chung KF, Adcock IM, COPDMAP (2015) Oxidative stress-induced mitochondrial dysfunction drives inflammation and airway smooth muscle remodeling in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol 136:769–780PubMedPubMedCentralCrossRef
137.
go back to reference Gershon AS, Warner L, Cascagnette P, Victor JC, To T (2011) Lifetime risk of developing chronic obstructive pulmonary disease: a longitudinal population study. Lancet 378:991–996PubMedCrossRef Gershon AS, Warner L, Cascagnette P, Victor JC, To T (2011) Lifetime risk of developing chronic obstructive pulmonary disease: a longitudinal population study. Lancet 378:991–996PubMedCrossRef
138.
go back to reference Makris D, Lazarou S, Alexandrakis M, Kourelis TV, Tzanakis N, Kyriakou D, Gourgoulianis KI (2008) Tc2 response at the onset of COPD exacerbations. Chest 134:483–488PubMedCrossRef Makris D, Lazarou S, Alexandrakis M, Kourelis TV, Tzanakis N, Kyriakou D, Gourgoulianis KI (2008) Tc2 response at the onset of COPD exacerbations. Chest 134:483–488PubMedCrossRef
139.
go back to reference Tsoumakidou M, Tzanakis N, Chrysofakis G, Kyriakou D, Siafakas NM (2005) Changes in sputum T-lymphocyte subpopulations at the onset of severe exacerbations of chronic obstructive pulmonary disease. Respir Med 99:572–579PubMedCrossRef Tsoumakidou M, Tzanakis N, Chrysofakis G, Kyriakou D, Siafakas NM (2005) Changes in sputum T-lymphocyte subpopulations at the onset of severe exacerbations of chronic obstructive pulmonary disease. Respir Med 99:572–579PubMedCrossRef
140.
go back to reference Qiu Y, Zhu J, Bandi V, Atmar RL, Hattotuwa K, Guntupalli KK, Jeffery PK (2003) Biopsy neutrophilia, chemokine and receptor gene expression in severe exacerbations of COPD. Am J Respir Crit Care Med 168:968–975PubMedCrossRef Qiu Y, Zhu J, Bandi V, Atmar RL, Hattotuwa K, Guntupalli KK, Jeffery PK (2003) Biopsy neutrophilia, chemokine and receptor gene expression in severe exacerbations of COPD. Am J Respir Crit Care Med 168:968–975PubMedCrossRef
141.
go back to reference Nagai A, West WW, Paul JL, Thurlbeck WM (1985) The National Institutes of Health Intermittent Positive-Pressure Breathing Trial: pathology studies. I. Interrelationship between morphologic lesions. Am Rev Respir Dis 132:937–945PubMed Nagai A, West WW, Paul JL, Thurlbeck WM (1985) The National Institutes of Health Intermittent Positive-Pressure Breathing Trial: pathology studies. I. Interrelationship between morphologic lesions. Am Rev Respir Dis 132:937–945PubMed
142.
go back to reference Nagai A, West WW, Thurlbeck WM (1985) The National Institutes of Health Intermittent Positive-Pressure Breathing Trial: pathology studies. II. Correlation between morphologic findings, clinical findings and evidence of expiratory air-flow obstruction. Am Rev Respir Dis 132:946–953PubMed Nagai A, West WW, Thurlbeck WM (1985) The National Institutes of Health Intermittent Positive-Pressure Breathing Trial: pathology studies. II. Correlation between morphologic findings, clinical findings and evidence of expiratory air-flow obstruction. Am Rev Respir Dis 132:946–953PubMed
143.
go back to reference Ritchie AI, Farne HA, Singanayagam A, Jackson DJ, Mallia P, Johnston SL (2015) Pathogenesis of viral infection in exacerbations of airway disease. Ann Am Thorac Soc 12(Suppl 2):S115–S132PubMed Ritchie AI, Farne HA, Singanayagam A, Jackson DJ, Mallia P, Johnston SL (2015) Pathogenesis of viral infection in exacerbations of airway disease. Ann Am Thorac Soc 12(Suppl 2):S115–S132PubMed
144.
go back to reference Hewitt R, Farne H, Ritchie A, Luke E, Johnston SL, Mallia P (2016) The role of viral infections in exacerbations of chronic obstructive pulmonary disease and asthma. Ther Adv Respir Dis 10(2):158–174PubMedCrossRef Hewitt R, Farne H, Ritchie A, Luke E, Johnston SL, Mallia P (2016) The role of viral infections in exacerbations of chronic obstructive pulmonary disease and asthma. Ther Adv Respir Dis 10(2):158–174PubMedCrossRef
145.
go back to reference Sajjan US, Jia Y, Newcomb DC, Bentley JK, Lukacs NW, LiPuma JJ, Hershenson MB (2006) H. influenzae potentiates airway epithelial cell responses to rhinovirus by increasing ICAM-1 and TLR3 expression. FASEB J 20:2121–2123PubMedCrossRef Sajjan US, Jia Y, Newcomb DC, Bentley JK, Lukacs NW, LiPuma JJ, Hershenson MB (2006) H. influenzae potentiates airway epithelial cell responses to rhinovirus by increasing ICAM-1 and TLR3 expression. FASEB J 20:2121–2123PubMedCrossRef
146.
go back to reference Marc MM, Korosec P, Kern I, Sok M, Ihan A, Kosnik M (2007) Lung tissue and tumour- infiltrating T lymphocytes in patients with non-small cell lung carcinoma and chronic obstructive pulmonary disease (COPD): moderate/severe versus mild stage of COPD. Scand J Immunol 66:694–702PubMedCrossRef Marc MM, Korosec P, Kern I, Sok M, Ihan A, Kosnik M (2007) Lung tissue and tumour- infiltrating T lymphocytes in patients with non-small cell lung carcinoma and chronic obstructive pulmonary disease (COPD): moderate/severe versus mild stage of COPD. Scand J Immunol 66:694–702PubMedCrossRef
147.
go back to reference Domagala-Kulawik J, Maskey-Warzecowska M (2009) T-regulatory cells in COPD or tumour environment. Eur Respir J 34:284, author reply 285–286 PubMedCrossRef Domagala-Kulawik J, Maskey-Warzecowska M (2009) T-regulatory cells in COPD or tumour environment. Eur Respir J 34:284, author reply 285–286 PubMedCrossRef
148.
go back to reference Perrot I, Blanchard D, Freymond N, Isaac S, Guibert B, Pachéco Y, Lebecque S (2007) Dendritic cells infiltrating human non-small cell lung cancer are blocked at immature stage. J Immunol 178:2763–2769PubMedCrossRef Perrot I, Blanchard D, Freymond N, Isaac S, Guibert B, Pachéco Y, Lebecque S (2007) Dendritic cells infiltrating human non-small cell lung cancer are blocked at immature stage. J Immunol 178:2763–2769PubMedCrossRef
149.
go back to reference de Boer WI, van Schadewijk A, Sont JK, Sharma HS, Stolk J, Hiemstra PS, van Krieken JH (1998) Transforming growth factor β1 and recruitment of macrophages and mast cells in airways in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 158:1951–1957PubMedCrossRef de Boer WI, van Schadewijk A, Sont JK, Sharma HS, Stolk J, Hiemstra PS, van Krieken JH (1998) Transforming growth factor β1 and recruitment of macrophages and mast cells in airways in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 158:1951–1957PubMedCrossRef
Metadata
Title
COPD immunopathology
Authors
Gaetano Caramori
Paolo Casolari
Adam Barczyk
Andrew L. Durham
Antonino Di Stefano
Ian Adcock
Publication date
01-07-2016
Publisher
Springer Berlin Heidelberg
Published in
Seminars in Immunopathology / Issue 4/2016
Print ISSN: 1863-2297
Electronic ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-016-0561-5

Other articles of this Issue 4/2016

Seminars in Immunopathology 4/2016 Go to the issue