Pathogenic triad in COPD: oxidative stress, protease-antiprotease imbalance, and inflammation

Bernard M Fischer, Elizabeth Pavlisko, Judith A Voynow, Bernard M Fischer, Elizabeth Pavlisko, Judith A Voynow

Abstract

Patients with chronic obstructive pulmonary disease (COPD) exhibit dominant features of chronic bronchitis, emphysema, and/or asthma, with a common phenotype of airflow obstruction. COPD pulmonary physiology reflects the sum of pathological changes in COPD, which can occur in large central airways, small peripheral airways, and the lung parenchyma. Quantitative or high-resolution computed tomography is used as a surrogate measure for assessment of disease progression. Different biological or molecular markers have been reported that reflect the mechanistic or pathogenic triad of inflammation, proteases, and oxidants and correspond to the different aspects of COPD histopathology. Similar to the pathogenic triad markers, genetic variations or polymorphisms have also been linked to COPD-associated inflammation, protease-antiprotease imbalance, and oxidative stress. Furthermore, in recent years, there have been reports identifying aging-associated mechanistic markers as downstream consequences of the pathogenic triad in the lungs from COPD patients. For this review, the authors have limited their discussion to a review of mechanistic markers and genetic variations and their association with COPD histopathology and disease status.

Keywords: apoptosis; bronchitis; chronic obstructive pulmonary disease; emphysema; senescence.

Figures

Figure 1
Figure 1
Histologic features of chronic bronchitis. (A) A section of bronchiole wall with luminal accumulation of mucous, goblet cell hyperplasia, basement membrane thickening (arrow), and scattered mononuclear inflammatory cells. (B) A bronchial wall with squamous metaplasia of the luminal epithelium (arrow head) and hyperplasia of the subepithelial seromucinous glands (arrow). Note: Hemotoxylin-eosin, original magnification ×200.
Figure 2
Figure 2
Histologic features of centrilobular emphysema. A section of lung tissue shows fragmented and “free-floating” alveolar septa (arrow) characteristic of emphysema. Note: Hemotoxylin-eosin, original magnification ×200.
Figure 3
Figure 3
Histologic features of small airways disease. A section of lung tissue shows accumulation of macrophages with smoker’s pigment (arrow) within and around a respiratory bronchiole (arrow head). Note: Hemotoxylin-eosin, original magnification ×200.
Figure 4
Figure 4
Pathogenic triad of COPD: oxidative stress, protease–antiprotease imbalance, and inflammation. Oxidative stress, protease–antiprotease imbalance and inflammation each are important in the pathogenesis of COPD; however, they constantly interact and may at times overlap with each other in the overall pathogenesis of COPD. As a consequence of oxidative stress, in particular cigarette smoking-induced oxidative stress, apoptosis, autophagy, and senescence are each potential lung cell fates. Senescent cells express a pro-inflammatory phenotype. Proteases, such as neutrophil elastase, have been shown in vitro to induce airway epithelial apoptosis, but this relationship has not yet been specifically demonstrated in human subjects. Listed in italics are the genetic polymorphisms that have been reported and discussed in this review, to be associated with COPD or emphysema in that area of the pathogenic triad.

References

    1. Wright JL, Churg A. Pathologic features of chronic obstructive pulmonary disease: diagnostic criteria and differential diagnosis. In: Fishman AP, Elias JA, Fishman JA, Grippi MA, Senior RM, Pack AI, editors. Fishman’s pulmonary diseases and disorders. 4th ed. Vol. 1. New York: McGraw Hill; 2008. pp. 693–705.
    1. Roggli V, Cagle P. Emphysema and chronic bronchitis. In: Tomashefski J, Cagle P, Farver C, Fraire A, editors. Dail and hammar’s pulmonary pathology. 3rd ed. New York: Springer; 2008. pp. 866–885.
    1. American Thoracic Society Standards for the diagnosis and care of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1995;152(5 Pt 2):S77–S121.
    1. Reid L. Measurement of the bronchial mucous gland layer: a diagnostic yardstick in chronic bronchitis. Thorax. 1960;15:132–141.
    1. Pratt P. Emphysema and chronic airways disease. In: Dail D, Hammar S, editors. Pulmonary pathology. 2nd ed. New York: Springer-Verlag; 1994. pp. 847–865.
    1. Thurlbeck W. Chronic airflow obstruction. In: Thurlbeck W, Churg A, editors. Pathology of the lung. 2nd ed. New York: Thieme Medical Publishers; 1995. pp. 739–825.
    1. Green L. Emphysema and diseases of large airways. In: Zander D, Farver C, editors. Pulmonary pathology. Philadelphia, PA: Churchill Livingstone/Elsevier; 2008. pp. 416–433.
    1. Hogg JC, Chu F, Utokaparch S, et al. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004;350:2645–2653.
    1. Hogg JC, Timens W. The pathology of chronic obstructive pulmonary disease. Annu Rev Pathol. 2009;4:435–459.
    1. Cagle P, Roggli V. Pathology of small airways. In: Tomashefski J, Farver C, Fraire A, Cagle P, editors. Dail and Hammar’s pulmonary pathology. 3rd ed. Vol. 1. New York: Springer; 2008. pp. 886–910.
    1. Grydeland TB, Dirksen A, Coxson HO, et al. Quantitative computed tomography measures of emphysema and airway wall thickness are related to respiratory symptoms. Am J Respir Crit Care Med. 2010;181:353–359.
    1. Patel BD, Coxson HO, Pillai SG, et al. Airway wall thickening and emphysema show independent familial aggregation in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2008;178:500–505.
    1. Rahman I, Adcock IM. Oxidative stress and redox regulation of lung inflammation in COPD. Eur Respir J. 2006;28:219–242.
    1. Rahman I, van Schadewijk AA, Crowther AJ, et al. 4-Hydroxy-2-nonenal, a specific lipid peroxidation product, is elevated in lungs of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2002;166:490–495.
    1. Kluchova Z, Petrasova D, Joppa P, Dorkova Z, Tkacova R. The association between oxidative stress and obstructive lung impairment in patients with COPD. Physiol Res. 2007;56:51–56.
    1. Montano M, Cisneros J, Ramirez-Venegas A, et al. Malondialdehyde and superoxide dismutase correlate with FEV(1) in patients with COPD associated with wood smoke exposure and tobacco smoking. Inhal Toxicol. 2010;22:868–874.
    1. Ghio AJ, Hilborn ED, Stonehuerner JG, et al. Particulate matter in cigarette smoke alters iron homeostasis to produce a biological effect. Am J Respir Crit Care Med. 2008;178:1130–1138.
    1. Fischer BM, Domowicz DA, Zheng S, et al. Neutrophil elastase increases airway epithelial nonheme iron levels. Clin Transl Sci. 2009;2:333–339.
    1. Britigan BE, Hayek MB, Doebbeling BN, Fick RB., Jr Transferrin and lactoferrin undergo proteolytic cleavage in the Pseudomonas aeruginosa-infected lungs of patients with cystic fibrosis. Infect Immun. 1993;61:5049–5055.
    1. Owen CA. Proteinases and oxidants as targets in the treatment of chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2005;2:373–385. discussion 394–375.
    1. Britigan BE, Edeker BL. Pseudomonas and neutrophil products modify transferrin and lactoferrin to create conditions that favor hydroxyl radical formation. J Clin Invest. 1991;88:1092–1102.
    1. Miller RA, Britigan BE. Protease-cleaved iron-transferrin augments oxidant-mediated endothelial cell injury via hydroxyl radical formation. J Clin Invest. 1995;95:2491–2500.
    1. DeMeo DL, Mariani T, Bhattacharya S, et al. Integration of genomic and genetic approaches implicates IREB2 as a COPD susceptibility gene. Am J Hum Genet. 2009;85:493–502.
    1. Comandini A, Marzano V, Curradi G, et al. Markers of anti-oxidant response in tobacco smoke exposed subjects: a data-mining review. Pulm Pharmacol Ther. 2010;23:482–492.
    1. Kensler TW, Wakabayashi N, Biswal S. Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol. 2007;47:89–116.
    1. Malhotra D, Thimmulappa R, Navas-Acien A, et al. Decline in NRF2-regulated antioxidants in chronic obstructive pulmonary disease lungs due to loss of its positive regulator, DJ-1. Am J Respir Crit Care Med. 2008;178:592–604.
    1. Kimpara T, Takeda A, Watanabe K, et al. Microsatellite polymorphism in the human heme oxygenase-1 gene promoter and its application in association studies with Alzheimer and Parkinson disease. Hum Genet. 1997;100:145–147.
    1. Yamada N, Yamaya M, Okinaga S, et al. Microsatellite polymorphism in the heme oxygenase-1 gene promoter is associated with susceptibility to emphysema. Am J Hum Genet. 2000;66:187–195.
    1. DeMeo DL, Hersh CP, Hoffman EA, et al. Genetic determinants of emphysema distribution in the national emphysema treatment trial. Am J Respir Crit Care Med. 2007;176:42–48.
    1. Nelson ME, O’Brien-Ladner AR, Wesselius LJ. Regional variation in iron and iron-binding proteins within the lungs of smokers. Am J Respire Crit Care Med. 1996;153(4 Pt 1):1353–1358.
    1. Young RP, Hopkins R, Black PN, et al. Functional variants of antioxidant genes in smokers with COPD and in those with normal lung function. Thorax. 2006;61:394–399.
    1. Juul K, Tybjaerg-Hansen A, Marklund S, Lange P, Nordestgaard BG. Genetically increased antioxidative protection and decreased chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2006;173:858–864.
    1. Dahl M, Bowler RP, Juul K, et al. Superoxide dismutase 3 polymorphism associated with reduced lung function in two large populations. Am J Respir Crit Care Med. 2008;178:906–912.
    1. Sorheim IC, DeMeo DL, Washko G, et al. Polymorphisms in the superoxide dismutase-3 gene are associated with emphysema in COPD. COPD. 2010;7:262–268.
    1. Deslee G, Woods JC, Moore C, et al. Oxidative damage to nucleic acids in severe emphysema. Chest. 2009;135:965–974.
    1. Harman D. Aging: a theory based on free radical and radiation chemistry. J Gerontol. 1956;11:298–300.
    1. Harman D. The free radical theory of aging. Antioxid Redox Signal. 2003;5:557–561.
    1. Finkel T, Holbrook NJ. Oxidants, oxidative stress and the biology of ageing. Nature. 2000;408:239–247.
    1. Ito K, Barnes PJ. COPD as a disease of accelerated lung aging. Chest. 2009;135:173–180.
    1. Aoshiba K, Nagai A. Senescence hypothesis for the pathogenetic mechanism of chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2009;6:596–601.
    1. MacNee W, Tuder RM. New paradigms in the pathogenesis of chronic obstructive pulmonary disease I. Proc Am Thorac Soc. 2009;6:527–531.
    1. Nyunoya T, Monick MM, Klingelhutz A, et al. Cigarette smoke induces cellular senescence. Am J Respir Cell Mol Biol. 2006;35:681–688.
    1. Tsuji T, Aoshiba K, Nagai A. Cigarette smoke induces senescence in alveolar epithelial cells. Am J Respir Cell Mol Biol. 2004;31:643–649.
    1. Tsuji T, Aoshiba K, Nagai A. Alveolar cell senescence in patients with pulmonary emphysema. Am J Respir Crit Care Med. 2006;174:886–893.
    1. Savale L, Chaouat A, Bastuji-Garin S, et al. Shortened telomeres in circulating leukocytes of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009;179:566–571.
    1. Mangino M, Brouilette S, Braund P, et al. A regulatory SNP of the BICD1 gene contributes to telomere length variation in humans. Hum Mol Genet. 2008;17:2518–2523.
    1. Kong X, Cho MH, Anderson W, et al. Genome-wide association study identifies BICD1 as a susceptibility gene for emphysema. Am J Respir Crit Care Med. 2011;183:43–49.
    1. Demedts IK, Demoor T, Bracke KR, Joos GF, Brusselle GG. Role of apoptosis in the pathogenesis of COPD and pulmonary emphysema. Respir Res. 2006;7:53.
    1. Morissette MC, Parent J, Milot J. Alveolar epithelial and endothelial cell apoptosis in emphysema: what we know and what we need to know. Int J Chron Obstruct Pulmon Dis. 2009;4:19–31.
    1. Kasahara Y, Tuder RM, Cool CD, et al. 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. 2001;163(3 Pt 1):737–744.
    1. Morissette MC, Vachon-Beaudoin G, Parent J, Chakir J, Milot J. Increased p53 level, Bax/Bcl-x(L) ratio, and TRAIL receptor expression in human emphysema. Am J Respir Crit Care Med. 2008;178:240–247.
    1. Takabatake N, Toriyama S, Igarashi A, et al. A novel polymorphism in CDC6 is associated with the decline in lung function of ex-smokers in COPD. Biochem Biophys Res Commun. 2009;381:554–559.
    1. Kim HP, Wang X, Chen ZH, et al. Autophagic proteins regulate cigarette smoke-induced apoptosis: protective role of heme oxygenase-1. Autophagy. 2008;4:887–895.
    1. Chen ZH, Kim HP, Sciurba FC, et al. Egr-1 regulates autophagy in cigarette smoke-induced chronic obstructive pulmonary disease. PLoS One. 2008;3:e3316.
    1. Karadag F, Karul AB, Cildag O, Yilmaz M, Ozcan H. Biomarkers of systemic inflammation in stable and exacerbation phases of COPD. Lung. 2008;186:403–409.
    1. Barnes PJ, Chowdhury B, Kharitonov SA, et al. Pulmonary biomarkers in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2006;174:6–14.
    1. Hacievliyagil SS, Gunen H, Mutlu LC, Karabulut AB, Temel I. Association between cytokines in induced sputum and severity of chronic obstructive pulmonary disease. Respir Med. 2006;100:846–854.
    1. O’Donnell R, Breen D, Wilson S, Djukanovic R. Inflammatory cells in the airways in COPD. Thorax. 2006;61:448–454.
    1. Cosio MG, Saetta M, Agusti A. Immunologic aspects of chronic obstructive pulmonary disease. N Engl J Med. 2009;360:2445–2454.
    1. O’Shaughnessy TC, Ansari TW, Barnes NC, Jeffery PK. Inflammation in bronchial biopsies of subjects with chronic bronchitis: inverse relationship of CD8+ T lymphocytes with FEV1. Am J Respir Crit Care Med. 1997;155:852–857.
    1. Saetta M, Baraldo S, Corbino L, et al. CD8+ve cells in the lungs of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1999;160:711–717.
    1. Saetta M, Di Stefano A, Turato G, et al. CD8+ T-lymphocytes in peripheral airways of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157(3 Pt 1):822–826.
    1. Saetta M, Mariani M, Panina-Bordignon P, et al. Increased expression of the chemokine receptor CXCR3 and its ligand CXCL10 in peripheral airways of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2002;165:1404–1409.
    1. Di Stefano A, Caramori G, Gnemmi I, et al. Association of increased CCL5 and CXCL7 chemokine expression with neutrophil activation in severe stable COPD. Thorax. 2009;64:968–975.
    1. Hizawa N, Makita H, Nasuhara Y, et al. Functional single nucleotide polymorphisms of the CCL5 gene and nonemphysematous phenotype in COPD patients. Eur Respir J. 2008;32:372–378.
    1. Sethi S, Mallia P, Johnston SL. New paradigms in the pathogenesis of chronic obstructive pulmonary disease II. Proc Am Thorac Soc. 2009;6:532–534.
    1. Qiu Y, Zhu J, Bandi V, et al. Biopsy neutrophilia, neutrophil chemokine and receptor gene expression in severe exacerbations of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2003;168:968–975.
    1. Takabatake N, Shibata Y, Abe S, et al. A single nucleotide polymorphism in the CCL1 gene predicts acute exacerbations in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2006;174:875–885.
    1. Campisi J, d’Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol. 2007;8:729–740.
    1. Tsuji T, Aoshiba K, Nagai A. Alveolar cell senescence exacerbates pulmonary inflammation in patients with chronic obstructive pulmonary disease. Respiration. 2010;80:59–70.
    1. Rajendrasozhan S, Yang SR, Kinnula VL, Rahman I. SIRT1, an antiinflammatory and antiaging protein, is decreased in lungs of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2008;177:861–870.
    1. Churg A, Wright JL. Proteases and emphysema. Curr Opin Pulm Med. 2005;11:153–159.
    1. Cavarra E, Lucattelli M, Gambelli F, et al. Human SLPI inactivation after cigarette smoke exposure in a new in vivo model of pulmonary oxidative stress. Am J Physiol Lung Cell Mol Physiol. 2001;281:L412–L417.
    1. Shapiro SD. Proteinases in chronic obstructive pulmonary disease. Biochem Soc Trans. 2002;30:98–102.
    1. Hunninghake GW, Davidson JM, Rennard S, et al. Elastin fragments attract macrophage precursors to diseased sites in pulmonary emphysema. Science. 1981;212:925–927.
    1. Weathington NM, van Houwelingen AH, Noerager BD, et al. A novel peptide CXCR ligand derived from extracellular matrix degradation during airway inflammation. Nat Med. 2006;12:317–323.
    1. Hautamaki RD, Kobayashi DK, Senior RM, Shapiro SD. Requirement for macrophage elastase for cigarette smoke-induced emphysema in mice. Science. 1997;277:2002–2004.
    1. Hunninghake GM, Cho MH, Tesfaigzi Y, et al. MMP12, lung function, and COPD in high-risk populations. N Engl J Med. 2009;361:2599–2608.
    1. Ito I, Nagai S, Handa T, et al. Matrix metalloproteinase-9 promoter polymorphism associated with upper lung dominant emphysema. Am J Respir Crit Care Med. 2005;172:1378–1382.
    1. Minematsu N, Nakamura H, Tateno H, Nakajima T, Yamaguchi K. Genetic polymorphism in matrix metalloproteinase-9 and pulmonary emphysema. Biochem Biophys Res Commun. 2001;289:116–119.
    1. Demeo DL, Mariani TJ, Lange C, et al. The SERPINE2 gene is associated with chronic obstructive pulmonary disease. Am J Hum Genet. 2006;78:253–264.
    1. Zhu G, Warren L, Aponte J, et al. The SERPINE2 gene is associated with chronic obstructive pulmonary disease in two large populations. Am J Respir Crit Care Med. 2007;176:167–173.
    1. Suzuki T, Moraes TJ, Vachon E, et al. Proteinase-activated receptor-1 mediates elastase-induced apoptosis of human lung epithelial cells. Am J Respir Cell Mol Biol. 2005;33:231–247.

Source: PubMed

3
Tilaa