1,031
Views
39
CrossRef citations to date
0
Altmetric
BASIC REVIEWS

Current Perspectives of Oxidative Stress and its Measurement in Chronic Obstructive Pulmonary Disease

&
Pages 291-306 | Published online: 30 Jul 2010

ABSTRACT

Cigarette smoking, the principal aetiology of chronic obstructive pulmonary disease (COPD) in the developed countries, delivers and generates oxidative stress within the lungs. This imbalance of oxidant burden and antioxidant capacity has been implicated as an important contributing factor in the pathogenesis of COPD. Oxidative processes and free radical generation orchestrate the inflammation, mucous gland hyperplasia, and apoptosis of the airway lining epithelium which characterises COPD. Pivotal oxidative stress/pro-inflammatory molecules include reactive oxygen species such as the superoxides and hydroxyl radicals, pro-inflammatory cytokines including leukotrienes, interleukins, tumour necrosis factor alpha, and activated transcriptional factors such as nuclear factor kappa-B and activator protein 1. The lung has a large reserve of antioxidant agents such as glutathione and superoxide dismutase to counter oxidants. However, smoking also causes the depletion of antioxidants, which further contributes to oxidative tissue damage. The downregulation of antioxidant pathways has also been associated with acute exacerbations of COPD. The delivery of redox-protective antioxidants may have preventative and therapeutic potential of COPD. Although these observations have yet to translate into common clinical practice, preliminary clinical trials and studies of animal models have shown that interventions to counter this oxidative imbalance may have potential to better manage COPD. There is, thus, a need for the ability to monitor such interventions and exhaled breath condensate is rapidly emerging as a novel and noninvasive approach in the sampling of airway epithelial lining fluid which could be used for repeated analysis of oxidative stress and inflammation in the lungs.

INTRODUCTION

Chronic obstructive pulmonary disease (COPD) is characterized by chronic airway inflammation, irreversible airflow limitation, and emphysema (Citation1). COPD is predicted to become the third leading cause of death and the fourth commonest cause of disability in the world by 2020 (Citation2). In the developed countries, cigarette smoking is the principal aetiology of COPD and is believed to account for approximately 90% of the cases (Citation3). There is a strong correlation between oxidative stress from cigarette smoking with the pathogenesis of COPD (Citation4, 5) as smokers have measurably elevated oxidants and depleted antioxidants in their exhaled breath and sputum samples (Citation6, 7) (). In the developing countries, a high prevalence of COPD have been documented in nonsmokers and never smokers (Citation8, 9). Risk factors of COPD include indoor air pollution from biomass fuel, pulmonary tuberculosis, chronic asthma, and socioeconomic status (Citation10). Occupational exposure of various compounds such as coal dust and asbestos can severely compromise lung function and may be risk factors for COPD (Citation10). This review will provide a basic overview of oxidative stress and its role in generating an oxidant/antioxidant imbalance in the lungs, contributing to the pathogenesis of COPD and summarize the potential of the exhaled breath analysis, a noninvasive method of airway sampling for assessing airway oxidative stress.

Figure 1. Schematic representation of oxidative stress. (a) In a healthy individual, there is an abundant of antioxidant capacities in the lungs to counteract inhaled oxidants from air pollution or acute exposures to cigarette smoke. (b) However, chronic smokers experience a reduced antioxidants and increased oxidant burden delivered from the cigarettes. At an early stage, the oxidant burden is still compensated by the antioxidant capacity but eventually (c) the oxidant burden overwhelms the body's antioxidant protection mechanism, leading to oxidative stress and the development of COPD.

Figure 1.  Schematic representation of oxidative stress. (a) In a healthy individual, there is an abundant of antioxidant capacities in the lungs to counteract inhaled oxidants from air pollution or acute exposures to cigarette smoke. (b) However, chronic smokers experience a reduced antioxidants and increased oxidant burden delivered from the cigarettes. At an early stage, the oxidant burden is still compensated by the antioxidant capacity but eventually (c) the oxidant burden overwhelms the body's antioxidant protection mechanism, leading to oxidative stress and the development of COPD.

OXIDANT GENERATION AND CIGARETTE SMOKING

Oxidative stress is the imbalance between the production of oxidants and the body's ability to detoxify the reactive intermediates or repair the resulting damage (Citation11). Oxidant generation is part of the normal cellular metabolism and is critical for cell homeostasis. Inflammatory cells such as macrophages, neutrophils, and eosinophils are the most important endogenous generators of oxidants ().

Table 1. Endogenous and exogenous sources of oxidants

More than 90% of COPD patients have a long-term cigarette smoking history. Cigarette smoke exposes the lung to extreme levels of oxidative stress. In the gaseous phase, cigarette smoke contains approximately >1015 reactive molecules per puff, including oxidants such as O2 and nitric oxides and peroxynitrite. The tar phase of cigarette smoke contains organic radicals such as long-lived semiquinone radicals, hydroxyl radicals, and H2O2. Side stream cigarette smoke contains more than 1017 reactive molecules per puff, including carbon monoxide, nicotine, ammonia, formaldehyde, and other carcinogenic organic molecules (Citation12). Cigarette smoke can induce increased oxidant burden (Citation13) and cause irreversible changes to the antioxidant protective effects in the airways (Citation14). Furthermore, the smoke-derived oxidants damage airway epithelial cells inducing direct injury to membrane lipids, protein, carbohydrates and DNA, leading to chronic inflammation (Citation15) ().

Figure 2. Consequences of airway damage by oxidative stress. Oxidative stress causes airway damages at three levels: airway changes, cellular changes, and biochemical changes. These factors contribute to lung injury and inflammation, which leads to the development of COPD. The ongoing oxidant injuries and inflammation in COPD feeds back to the increased airway damage, restarting the vicious cycle.

Figure 2.  Consequences of airway damage by oxidative stress. Oxidative stress causes airway damages at three levels: airway changes, cellular changes, and biochemical changes. These factors contribute to lung injury and inflammation, which leads to the development of COPD. The ongoing oxidant injuries and inflammation in COPD feeds back to the increased airway damage, restarting the vicious cycle.

Chronic inflammation is a prominent feature in COPD demonstrated by the increased number of activated neutrophils (Citation16), macrophages (Citation17), and inflammatory mediators in the airways (Citation7). The combination of oxidative stress and chronic inflammation accounts for the increased free radicals in the airways and further enhances pro-inflammatory gene expression, inflammatory protein release, and inactivation of anti-proteases, leading to a vicious cycle of oxidative injury and inflammatory cell recruitment () (Citation7, Citation18, Citation19).

Figure 3. Vicious cycle of oxidative stress and inflammation (adapted from Lee 2009) (154). Oxidative stress generated by cigarette smoking increases ROS production in the bronchial mucosa, leading to the recruitment of inflammatory cells, which further increases oxidative stress. These phenomena cause lung injuries, which further enhances lung tissue inflammation and the generation of more ROS.

Figure 3.  Vicious cycle of oxidative stress and inflammation (adapted from Lee 2009) (154). Oxidative stress generated by cigarette smoking increases ROS production in the bronchial mucosa, leading to the recruitment of inflammatory cells, which further increases oxidative stress. These phenomena cause lung injuries, which further enhances lung tissue inflammation and the generation of more ROS.

ROLE OF OXIDATIVE STRESS IN COPD

There is strong evidence to suggest an association between the increased oxidant burden and the development of COPD. Oxidative processes and free radical generation alter gene transcription and orchestrate inflammation, mucous gland hyperplasia, and apoptosis of the airway lining epithelium, all of which characterizes COPD.

Altered gene transcription

Transcription of genes starts with the histone acetyl transferase (HAT) controlled unravelling of DNA from histones to allow access to transcription factors. Oxidative stress activates HAT activity in respiratory epithelial cells, leading to inflammatory gene transcription (Citation20). Reactive oxidant species (ROS) may indirectly cause an upregulation of HAT activity via activating NF-κB, AP-1, and other redox-sensitive transcriptional factors (Citation21).

The oxidant burden within the airway epithelium and alveolar macrophages may generate cytokines which increase recruitment of pro-inflammatory cells (Citation22). The development of COPD may be closely related to the imbalance of pro-inflammatory gene upregulation (Citation23) and depleting the antioxidant protective mechanism in the lungs can induce an increased oxidant burden in the airways.

Mucus gland hypersecretion

Two coexisting mucosal phenomena occur in COPD: impaired mucociliary clearance and increased secretion. Endogenous and exogenous oxidants such as superoxides and H2O2 may impair mucociliary clearance by decreasing respiratory epithelial cilia beat frequency as early as 15 minutes after exposure (Citation24). On the other hand, various ROS and inflammatory cytokines have been reported to directly or indirectly stimulate mucus hypersecretion (Citation25, 26). Specifically, TNF-α (Citation27), IL-1β (Citation28), and lipo-polysacchride (Citation29) have been shown to activate mucin synthesis by upregulation and expression of the mucin genes (Citation26).

Apoptosis

COPD is characterized by progressive destruction of lung parenchyma, leading to irreversible air obstruction. Apoptosis, or programmed cell death, is a highly regulated mechanism that allows elimination of unwanted, damaged, or infected cells which may occur prematurely (Citation30) (), leading to apoptosis of structural cells in the lungs which may possibly be an upstream event in the pathogenesis of COPD (Citation30). Current evidence indicates an increased rate of apoptosis in endothelial cells (Citation31, 32) alveolar epithelial cells (Citation33, 34), lung mesenchymal cells (Citation35), and inflammatory cells (Citation32, Citation35) in COPD and emphysema patients. Apoptosis within the respiratory tract is also directly related to the decline of lung function in COPD patients (Citation36).

Table 2. Mechanisms of controlled apoptosis (Recreated from Demedts 2006; Hodge 2005) (30, 35)

Inflammation

Increased airway inflammation is a characteristic for many respiratory diseases including influenza infection, asthma, and COPD; however, the patterns of inflammation differ. Sputum samples of asthmatic patients demonstrate an eosinophil dominated pattern due to the nature of asthma being a hyper-reactivity of the immune system toward allergens (Citation37). The COPD disease process demonstrates a predominantly neutrophilic pattern (Citation16), which is inversely correlated with lung function (FEV1) (Citation38), indicating that neutrophilic inflammation is closely linked to increased airway obstruction. Oxidative stress may directly recruit inflammatory cells and cytokines to the lungs and set up a positive feedback loop that perpetuates further inflammation and tissue destruction by the infiltration of excess immune cells (Citation39).

Quantification of these inflammatory and oxidative stress markers in COPD has thus become a focus of research. Selective biomarkers may assist in making accurate measurements of disease state/severity and provide potential pharmaceutical targets to better manage COPD.

OXIDATIVE STRESS AND INFLAMMATION MARKERS IN COPD

Oxidative stress markers reflect the level of free oxidants/ROS generating capacity in the airways and lungs. Inflammatory markers reflect the level of inflammation in the airways/lungs. They are generally molecules that directly promote local inflammation or indirectly recruits and attracts inflammatory cells.

Markers of oxidative stress

Hydrogen peroxide (H2O2)

H2O2 is a reactive oxygen species that can produce the highly reactive oxidants and free radicals including hydrochlorous acid (formed by myeloperoxidase) and hydrobromous acid. An example of which is the transition metal (Fe3+, Cu2+) catalyzed Haber–Weiss reaction: [1]

H2O2 is also a product of many redox-active metal catalyzed reactions, leading to more oxidant generating potential:

  • Ascorbic Acid + O2 → Ascorbyl radical, ·OH, H2O2

  • NAD(P)H + O2 → NAD(P)·, ·O2, ·OH, H2O2

  • Catecholamines + O2 → ·O2, ·OH, H2O2

  • RSH (thiols) + O2 → O2, ·OH, H2O2, RS· (thiyl radical).

H2O2 is a good indicator of COPD disease state because of the significant difference in exhaled breath/sputum concentration between healthy subjects, COPD patients, and patients with acute exacerbated conditions (Citation40–42).

pH

pH is a robust and reproducible method of sampling airway acidity but it is only an indirect and nonspecific measure for detecting oxidative stress (Citation43). A decreased pH in response to oxidative stress is a result of inflammation and the decline in the action of Na+/H+ exchanger on the cellular membrane (Citation44). The airway pH is inversely correlated with respiratory disease activity in conditions such as asthma, COPD, and acute exacerbations (Citation45). The stability and bioactivity of many reactive nitrogen and oxygen species are pH dependent. For example, airway acidity contributes to an increased NO concentration since it promotes eosinophil necrosis and causes conversion of endogenous nitrate to NO (Citation21).

8-isoprostane

The detection and measurement of lipid peroxidation products such as prostaglandin also serve as a marker of oxidant burden. In particular, 8-isoprostane, a relatively stable marker at physiological temperature, is a prostaglandin analog produced by free-radical-catalyzed peroxidation of arachidonic acid (Citation46). 8-isoprostane levels were significantly elevated in breath samples of patients with COPD, irrespective of lung function impairment (Citation47). Furthermore, it has been suggested that 8-isoprostane level may reflect the extent of lung emphysema in COPD patients (Citation47).

Malonyldialdehyde (MDA)

Serum MDA, a product of cell membrane lipid peroxidation, is a marker of oxidative stress, and when measured in COPD and healthy control subjects, serum MDA levels were highest in COPD patients with exacerbation, followed by stable COPD patients, followed by healthy control participants (Citation48, 49).

Nitric oxide and reactive nitrogen species

Nitric oxide (NO), nitrite (NO2), and nitrate (NO3) have important physiological roles in the respiratory tract including vascular regulation, neurotransmission, host defence, and cytotoxicity. Reactive nitrogen species (RNS) may contribute to oxidative injury via the interaction with reactive oxygen radicals, giving rise to reactive nitrogen intermediates (Citation50), RNS has also been linked to the development of COPD (Citation51). NO is highly unstable and can react with superoxide anions (O2) to form peroxynitrate, an oxidizing agent that contributes to lipid peroxidation, hydroxylation, and nitration of amino acids. However, despite its ability to induce oxidative stress, the direct involvement of RNS in the development of COPD remains controversial (Citation52–55).

Peroxynitrites 3-nitrotyrosine (3-NT)

Peroxynitrites (PN) are powerful and cytotoxic-reactive intermediates in biologic systems and are able to react with multitude of organic compounds to generate oxidants. PN alter the function of many antioxidant proteins such as superoxide dismutase (SOD), glutathione s-transferase (GST), and metalloproteinase (Citation52, Citation56). Indirect measurement of PN in COPD EBC indicated elevated PN compared to control samples, with a strong correlation of PN activity and lung function (FEV1) (Citation52). However, direct quantification of PN is extremely difficult due to their short half-life (<1 s in pH 7.4) (Citation52). RNS-induced oxidative stress is thus quantified by the measurement of NO, nitrite/nitrate, and nitro-tyrosine as a footprint of PN formation.

3-nitrotyrosine (3-NT) is derived from the nitration of the amino acid tyrosine or tyrosine residues within proteins. Using high-performance liquid chromatography (HPLC), 3NT was found to be upregulated in COPD and asthmatic patients (Citation57, 58). A higher number of 3-NT positive cells were also found in the submucosa of severe COPD patients as compared with patients with mild/moderate COPD, healthy smokers, and healthy nonsmokers (Citation59).

Markers of inflammation

Pro-inflammatory transcription precursors

Nuclear factor κB (NF-κB) is a protein complex that controls transcription of DNA as well as playing a key role in cellular immune responses to potentially damaging stimuli such as stress, cytokines, free radicals, oxidized LDL, and bacterial or viral antigens. Activator protein-1 (AP-1) is also a transcription factor protein that regulates gene expression in response to a variety of stimuli including cytokines, stress, oxidants, and infection. AP-1 controls cellular processes including the differentiation, proliferation, and apoptosis of inflammatory cells. NF-κB and AP-1 are upregulated by oxidants, leading to an increase in airway inflammatory cytokines, direct recruitment of inflammatory cells, resulting in lung tissue injuring and causing a further inflammatory response (Citation7, Citation18, Citation41, Citation45, Citation60).

Leukotrienes

Leukotrienes are lipid-derived mediators of the immune system produced endogenously from arachidonic acid by the enzyme 5-lipoxygenase, which plays a key role in the inflammation of asthma and COPD (Citation21). Leukotriene B4 (LTB4) is chemotactic to neutrophils and is found in elevated concentrations in exhaled breath (Citation61) and induced sputum (Citation21) in COPD patients and smokers as compared with nonsmokers. Furthermore, it has been shown that LTB4 concentrations are significantly correlated with interleukin-6 (Citation61).

The cysteinyl (Cys)-leukotrienes—LTC4, LTD4, and LTE4—are responsible for airway smooth muscle contraction, increasing vascular permeability and stimulate mucus secretion, all of which would induce an acute asthmatic attack (Citation62). The use of leukotriene antagonists in the treatment of asthma is commonplace; however, there has not been sufficient data to suggest a benefit of leukotriene antagonism in the treatment of COPD.

Interleukins

Interleukins (ILs) are a group of pro-inflammatory cytokines (signalling molecules) that are expressed by leukocytes. Several ILs have been associated with COPD, including IL-1β (Citation63), IL-6 (Citation61), IL-8 (Citation64), IL-10 (Citation65), among others. Their primary functions are to stimulate inflammatory cell chemotaxis and to induce the maturation and proliferation of immune cells including neutrophils, macrophages, T-helper cells, B-cells, and natural killer cells. IL-6 levels are significantly higher in smokers than control subjects (Citation61) and IL-8 levels are significantly higher in induced sputum of COPD patients (Citation66), suggesting a direct relationship between airway IL activity and local inflammation and tissue damage.

Tumor necrosis factor-α

Tumor necrosis factor-α (TNF-α) is a cytokine that stimulates acute phase reaction and is involved in systemic inflammation. The primary function of TNF is the regulation of immune cells but it is also capable of inducing apoptosis. TNF-α is produced mainly by macrophages, which are increased in smokers and COPD patients. Exhaled breath condensate and induced sputum studies have shown an elevated TNF-α concentration in smokers as compared to nonsmokers (Citation66, 67). Correlations between TNF-α have been found in COPD patients with other mediators of inflammation such as IL-1β, LTB4, sputum neutrophils, and sputum macrophages (Citation68). However, there was considerable variation in the strength of the correlations.

Transforming growth factor-β

Transforming growth factor-β (TGF-β) is a fibrogenic and immune-modulatory cytokine that controls a range of cellular functions including proliferation, differentiation, apoptosis, and recruitment of inflammatory cells when activated (Citation69). TGF-β has been shown to perpetuate the pathogenesis of asthma by airway remodelling (Citation70) and COPD through its induction of inflammation (Citation69).

Volatile organic compounds

Volatile organic compounds (VOCs) refer to chemical compounds that have significant vapor pressure and low water solubility. VOCs can include both naturally occurring and man-made chemical compounds that can affect the environment and human health. These compounds are generally not acutely toxic but have chronic effects in which the mechanism have yet to be fully elucidated.

In the area of respiratory medicine and COPD, exhaled breath analysis has identified six potential VOCs (isoprene, C16 hydrocarbon, 4,7-dimethy undecane, 2,6-dimethy heptane, 4-methyloctane, and hexadecane) having early diagnostic potential for COPD (Citation71). Additionally, VOC “fingerprints” from exhaled breath have been documented to be able to discriminate between COPD and healthy control participants (Citation72). This finding furthers the potential for exhaled VOC as a tool for early detection and diagnosis of COPD.

Other inflammatory markers

Vascular endothelial growth factor (VEGF) has been implicated in the pulmonary remodelling of chronic progressive airflow limitation of COPD. Although VEGF is most potent as a regulator of angiogenesis, VEGF expression is increased in chronic inflammation and has been implicated in the pathogenesis of emphysema through apoptotic and oxidative stress mechanisms (Citation73–75). The median VEGF concentration is also significantly elevated in induced sputum of COPD patients compared to nonsmokers and asymptomatic smokers (Citation66).

ANTIOXIDANTS OF THE LUNGS

Antioxidants are agents that decrease steady-state ROS and protect cellular macromolecules from oxidative modification (Citation76). A classic antioxidant rapidly reacts with ROS, producing less-reactive species. Broadly speaking, there are two categories of endogenous antioxidants: enzymatic and nonenzymatic antioxidants (Citation77). Antioxidant enzymes include SOD, catalase, glutathione (GSH) peroxidases, GSH S-transferase, peroxiredoxin, and the heme-oxygenase-I system. Nonenzymatic antioxidants include GSH, ascorbate, urate, alpha-tocopherol, bilirubin, carbon monoxide, ferritin, and lipoic acid.

Enzymatic antioxidants

Superoxide dismutases family

SODs are a class of enzymatic antioxidants that catalyze the dismutation of superoxide into oxygen and hydrogen peroxide. The chemical reaction can be written as follows:

  • (Transitional metal(n+1)+ − SOD) + O2 → (transitional metaln+ − SOD) + O2

  • (Transitional metaln+ − SOD) + O2 + 2H+ → (transitional metal(n+1)+ − SOD) + H2O2.

The are three forms of superoxide dismutases in humans, Cu/ZnSOD and MnSOD are primarily localized in lung cells and extracellular SOD (EcSOD) is mainly in the extracellular space of the lung (Citation78).

Cu/ZnSOD is the most abundant superoxide dismutase in lung tissue, and is especially concentrated in the ciliated epithelial cells to combat inhaled oxidants (Citation77). An overexpression of transgenic CuZnSOD can resist allergen-induced lung toxicity and pulmonaryoxygen toxicity (Citation79, 80). MnSOD is predominantly located in cells in airway walls and mitochondria of the lungs (Citation81). An over-expression of transgenic MnSOD in lungs of mice had protective effects toward hyperoxia (Citation82). MnSOD expression is sensitive to oxidative stress. The concentration of both bronchial/alveolar epithelium MnSOD is higher in COPD than in nonsmokers and MnSOD activity is increased by 50% in smokers as compared to nonsmokers (Citation83).

EcSOD is the major SOD in the extracellular fluid that exists in abundance in the lungs, the fluid lining the lung and in the vasculature (Citation78, Citation84, Citation85). EcSOD is capable of protecting the lung from free radical damage and in controlling inflammation (Citation86). There is strong evidence documenting the role of EcSOD in controlling inflammation and fibrosis of the lungs via reducing the expression of inflammatory cytokines TNF-α and MIP-2 (Citation39). Additionally, functional polymorphisms of the SOD gene correlate well with lung function and disease status (Citation87–91) ().

Table 3. Summary of genetic association studies of COPD with EcSOD (Reproduced from Oberley-Deegan et al. 2009) (78)

Glutathione (GSH) redox system

GSH is the most abundant intracellular thiol-based antioxidant that reacts with peroxides forming less-toxic disulfide products and is recycled by gluthathione-reductases (GR). GSH is more concentrated in the epithelial lining fluids and plays an important role in the maintenance of the redox balance in the lungs. In addition, GSH, under catalysis of GSH-S-transferase, is capable of rapidly detoxifying harmful components of tobacco smoke (Citation92). Glutathione peroxidases (GPx) also catalyze a variety of GSH reactions including the breakdown of H2O2 ():

Figure 4. Diagrammatic representation of the GSH redox system. The oxidant hydrogen peroxide (H2O2) generated from oxidative stress is neutralized by the glutathione redox system with the assistance of GSH-reductase and glutathione peroxidase.

Figure 4.  Diagrammatic representation of the GSH redox system. The oxidant hydrogen peroxide (H2O2) generated from oxidative stress is neutralized by the glutathione redox system with the assistance of GSH-reductase and glutathione peroxidase.

  • 2GSH + H2O2 → GS–SG + 2H2O.

The GSH recycle reaction catalyzed by GR occurs as follows:

  • GS – SG + NADPH + H+ → 2 GSH + NADP+.

GSH is sensitive to oxidative stress. Acute cigarette smoke exposure of alveolar epithelial cells in culture or to lung in animal models depleted GSH (Citation93, 94). However, GSH levels increased in the epithelial lining fluids of chronic cigarette smokers (Citation95), suggesting a transient upregulation of GSH in acute exposure to counteract the increased oxidants, but it is eventually overwhelmed by ongoing oxidative stress. Cigarette smoke also affects GPx, GR, and GSH-S-transferase, all of which are detoxifying enzymes involved in the GSH redox system in the lungs. These observations suggest that smokers are predisposed to oxidative injuries.

Peroxiredoxins and thioredoxins

Peroxiredoxins are a family of antioxidant enzymes that control cytokine-induced peroxide levels by using cysteine as the catalytic center rather than selenocysteine in the GPx enzymes (Citation96). Thioredoxins are proteins that act as antioxidants by facilitating (as a cofactor) the reduction of H2O2 and organic hydroperoxides. The antioxidant capacity of the peroxiredoxins (mainly the peroxiredoxin-6) has been assessed by attenuating the gene expression. A decreased expression of peroxiredoxins in rat lung epithelial cell line resulted in increased oxidant sensitivity and apoptosis (Citation97). Conversely, an increased expression resulted in decreased oxidative stress and increased survival in hyperoxia (Citation98, 99). Peroxiredoxins have increased expression in chronic oxidative diseases such as COPD (Citation96).

Catalase

Catalase is a common enzymatic antioxidant found in living organisms. It functions to catalyze the decomposition of H2O2 to water and oxygen:

  • 2H2O2 → 2H2O + O2.

Catalase is an extremely well-researched antioxidant whose imbalance has been linked to aging (Citation100), atherosclerosis (Citation101), among others. In terms of respiratory health, catalase has been demonstrated to be significantly downregulated in former smokers with COPD compared to never-smokers and former smokers without COPD (Citation102). Additionally, polymorphisms of the catalyse gene have been documented to be related to an imbalanced oxidant/antioxidant system in patients of COPD (Citation103).

Heme-oxygenase-1 (HO-1)

Heme-oxygenase (HO) is an enzyme that catalyzes the degradation of heme, an oxidant, and generates biliverdin-IXα, iron, and carbon monoxide (CO) (). These downstream products of heme catabolism mediate the antioxidant properties of HO. In the airways, HO-1 is expressed in the epithelium, smooth muscle, type II pneumocytes, and alveolar macrophages. HO-1 is induced by heme, hypoxia, hyperoxia, NO, endotoxin, and pro-inflammatory cytokines (Citation104). HO-1 expression is extremely sensitive to agents that cause oxidative stress and exhibits, in turn, powerful antioxidant and anti-inflammatory properties. HO-1 expression is upregulated in several pulmonary diseases, including COPD and asthma. The over-expression of HO-1, demonstrated in animal models, can be beneficial in several lung diseases including COPD (Citation105) ().

Figure 5. Integrated diagram of oxidative stress and the heme breakdown pathway by HO-1. Heme is a product of haemoglobin metabolism and is further broken down into carbon monoxide, iron, and biliverdin, which is then converted into bilirubin. Each of these downstream products of heme metabolism has antioxidant capacities that function at different levels. CO acts as anti-inflammatory, anti-apoptotic, anti-proliferative agent. The iron-storage protein ferritin has antioxidant actions that counteract oxidative stress, and bilirubin is also an antioxidant and anti-inflammatory molecule.

Figure 5.  Integrated diagram of oxidative stress and the heme breakdown pathway by HO-1. Heme is a product of haemoglobin metabolism and is further broken down into carbon monoxide, iron, and biliverdin, which is then converted into bilirubin. Each of these downstream products of heme metabolism has antioxidant capacities that function at different levels. CO acts as anti-inflammatory, anti-apoptotic, anti-proliferative agent. The iron-storage protein ferritin has antioxidant actions that counteract oxidative stress, and bilirubin is also an antioxidant and anti-inflammatory molecule.

Nonenzymatic antioxidants

HO-1 downstream products (bilirubin, carbon monoxide, ferritin)

HO-1 generates biliverdin, which is then converted to bilirubin, a potent endogenous antioxidant (Citation106). CO has numerous biological functions including anti-inflammatory, anti-apoptotic, anti-coagulation, and anti-proliferative effects (Citation53). CO can also inhibit smooth muscle proliferation, platelet aggregation, and modulate vascular tone by increasing cGMP levels (Citation104, Citation107, Citation108). Free iron produced by HO-1 is taken up by ferritin, a major intracellular iron storage protein that removes excess free iron, thus minimizing the generation of iron-catalyzed reactive oxygen species.

α-Tocopherol and ascorbate

Vitamins E (α-tocopherol) and C (ascorbate) are two of the dominant naturally occurring antioxidants in human. They function by binding with ROS to produce less reactive radicals that can be recycled by cellular reductases (Citation76). Oral vitamin A, C, E supplementations have protective effects upon oxidant-induced airway damage (Citation109, 110). A water soluble derivative of α-tocopherol is 6-hydroxy-2,5,7, 8-tetramethylchroman-2-carboxylic acid (Trolox®). Trolox® is used in biological/biomedical applications to reduce oxidative stress or damage. Because of the difficulties in measuring individual antioxidant components of a complex mixture, Trolox® equivalency antioxidant capacity (TEAC) is used as a benchmark for antioxidant capacity.

N-acetyl-L-cysteine

N-acetyl-L-cysteine (NAC) is a powerful antioxidant capable of removing several ROS such as hypochlorous acid, peroxides, hydroxyl radicals, and ONOO (Citation76, Citation111). Additionally, NAC stimulates GSH synthesis, enhances glutathione-S-transferase activity, and promotes liver detoxification. The therapeutic use of NAC in COPD has yield conflicting results, and while some studies have shown beneficial effects of NAC with regard to lung function (Citation112) and COPD exacerbations (Citation113–115), some clinical trials concluded that NAC was ineffective at preventing deterioration of lung function or preventing exacerbations (Citation116, 117).

Role of antioxidants in COPD

A relationship between dietary antioxidants, pulmonary function, and the development of COPD has been shown in several studies. Population surveys showed the inverse relationship between both dietary and serum vitamin C to FEV1 and chronic respiratory symptoms (Citation118, 119). A positive relationship was found between the levels of dietary vitamin C, vitamin E, selenium, and beta-carotene and FEV1 in healthy and COPD subjects (Citation120–123).

As a response to oxidative stress, there is a transient upregulation of protective antioxidant generation within the lungs including MnSOD, metallothionein, and GSH peroxidase (Citation124). However, in COPD patients, serum/airway antioxidant levels are depleted (Citation125, 126), suggesting that the increase of airway antioxidants in response to oxidative stress is a temporary phenomenon and this protective mechanism can be overwhelmed by chronic exposure of airway and lungs to oxidative stress, for example in chronic smokers.

EXACERBATIONS OF COPD

Exacerbations of COPD are characterized by an acute increase in symptoms, associated with a deterioration in lung function, resulting in poor health and high mortality. Causes of exacerbations include bacterial/viral infection, continued oxidant damage, air pollution, comorbidities of COPD, pulmonary hypertension, and cor pulmonale (Citation127). Advanced diagnostic techniques have demonstrated rhinovirus (Citation128) and seasonal influenza (Citation129) as a common cause of COPD exacerbation by directly infecting the upper-respiratory and lower-respiratory tract. Bacterial colonization is often reported in patients with COPD and associated with frequency of exacerbations (Citation130). Exacerbations of COPD reflect a worsening of airway and lung oxidative stress caused by acute-on-chronic inflammation that overwhelms the protective oxidant-scavenging activity. The increased oxidative state can be caused by intrinsic factors due to worsening of the oxidant burden and depleted antioxidant capacity or can be triggered by extrinsic factors such as air pollution and acute bacterial/viral infections (Citation7, Citation126).

COPD subjects with exacerbations exhibit an increased oxidative stress associated with elevated IL-8 and neutrophil activity in the distal airspaces (Citation7), in combination with depleted antioxidants such as decreased plasma TEAC and GSH levels in patients with acute exacerbations of COPD (Citation7, Citation126, Citation131). Worsening of oxidative stress also induces the transcription of various pro-inflammatory factors such as NF-κB (Citation132), AP-1 (Citation7), which translates to inflammation and a decline in lung function. A compensatory mechanism in response is the transient upregulation of antioxidant genes, such as the HO-1 pathway, but fails to restore oxidant/antioxidant balance completely (Citation124–126).

A rational approach to the treatment of COPD exacerbations should thus focus on its three most common causes—oxidative stress, viruses, and bacterial infections. These factors amplify the innate inflammatory response, triggering increased oxidant production and might inhibit anti-inflammatory responses. Therefore, antioxidants may have an important role in the treatment of COPD exacerbations.

THERAPEUTIC POTENTIAL OF OXIDANT INHIBITION AND ANTIOXIDANT IN COPD

Various approaches have been taken to redress the oxidant/antioxidant imbalance with the intervention of ameliorating lung function decline and to better manage COPD. Two potential therapeutic options are the reduction of oxidant burden and the delivery of antioxidants, with their therapeutic effects indicated in countering oxidative stress in the airways (Citation115, Citation133, Citation134) ().

Table 4. Recent clinical trials for the efficacy of antioxidants in pulmonary disease and in the general population

Direct antioxidants

Dietary antioxidant supplementation is one of the simplest approaches in delivering antioxidants to the body. The most common dietary antioxidant supplementations are vitamins A, C, E including β-carotene, through tablets or vegetables and fruits. A diet high in vegetables and fruits has an inverse relationship with COPD risk factors (Citation135), and supplementation with vitamins C and E is associated with an improvement of oxidant/antioxidant imbalance in COPD (Citation110). Supplementation of β-Carotene may also protect against FEV1 decline in the general population (Citation109). Oral administration of NAC in the management of COPD has yielded some positive results as NAC may improve physical performance and exercise tolerance (Citation112), reduce odds of exacerbation (Citation136), lower oxidant markers, and improve clinical outcome in managing acute exacerbation (Citation137). Some studies have also shown that the use of NAC could reduce rate of hospitalization of COPD patients by 30% with no decline of FEV1 (Citation117), although not all studies have yielded beneficial results. The use of intra-tracheal injection of MnSOD in an animal model revealed protective effect in otherwise healthy lungs (Citation138).

Another possible approach of antioxidant therapy is to deliver antioxidants to the lungs. GSH is a potential antioxidant in the management of COPD. A range of studies have suggested that inhaled GSH may be beneficial in other lung diseases such as cystic fibrosis (Citation139, 140) and idiopathic pulmonary fibrosis (Citation141, 142).

Indirect antioxidants

Indirect antioxidants include the use of antioxidant analogs that are converted into antioxidants in vivo or the use of medications to modulate the oxidant/antioxidant level in the body. N-acystelyn (NAL), a salt of NAC, is a mucolytic and antioxidant compound. It can be aerosolized into the lungs to enhance intracellular GSH in alveolar epithelial cells and inhibit the formation of ROS (Citation143). All-trans retinoic acid, an acidic form of vitamin A, can modulate protease/anti-protease balance in the lungs (Citation144).

Oxidant inhibitors

Inhibition of superoxide generation via phosphodiesterase 4 (PDE4) inhibitors is a novel method to control oxidative stress. PDE4 inhibitors promote intracellular levels of cyclic adenosine monophosphate (cAMP), which has a broad anti-inflammatory effect. Oral intake of PDE4 inhibition produced significant improvement in lung function of COPD patients (Citation145); however, an inhaled PDE4 inhibitor showed no improvement in lung function of COPD symptoms (Citation146). Other potential pharmaceutical targets include redox sensitive molecules such as NF-κB, AP-1, or inflammatory cytokines such as TNF-α and monocyte inflammatory protein (MIP)-1β.

QUANTIFYING OXIDATIVE STRESS AND ANTIOXIDANTS IN THE AIRWAY AND LUNGS

The accepted method of investigating the respiratory system has depended upon invasive approaches such as induced sputum, broncho-alveolar lavage, or lung biopsy. However, the invasive nature of these procedures made it sub-optimal for as tools of disease diagnosis or long-term monitoring. Thus, there has been an increasing interest in using exhaled breath as a simple, noninvasive means to repeatedly sample and monitor the lower respiratory tract (Citation63).

Exhaled breath condensate

Exhaled breath contains aerosol particles and proteins that originate in the lower respiratory tract (Citation147). Proteomic comparisons between frozen aerosol particles and saliva samples demonstrate the presence of extra protein in aerosol particles that are not found in saliva, suggesting that nonvolatile substances such as proteins from the lower respiratory tract can be transported in the form of aerosols in exhaled breath (Citation63).

Exhaled breath can be collected in the gaseous phase or the liquid phase. The gaseous phase comprises products of oxidative stress and antioxidants such as CO, NO, and volatile organic compounds (Citation148). The liquid phase, or the exhaled breath condensate (EBC), comprises nonvolatile mediators and inflammatory markers such as hydrogen peroxide, nitrites/nitrates, nitrotyrosine, adenosine, vasoactive amines, ammonia, eicosanoids, peptides, cytokines, and other larger molecules (Citation149, 150).

EBC collection method

EBC is collected by the condensation of expired air through a cooling system. Successful collection has been reported with many devices, but different designs may contribute to biomarker variability (Citation151). There is intersubject variability of EBC volume generated and minute ventilation remains the major determinant of the amount of condensate obtained over time (Citation147, Citation149).

EBC samples airway lining fluid (Citation65, Citation152) from the upper respiratory tract (nasopharynx, trachea), lower respiratory tract (bronchi and alveoli), and the ambient environment and the oral cavity (Citation149). Therefore, prevention of contamination is an important step in designing and sampling of EBC. Saliva is a major source of contamination, and salivary amylase can be tested in EBC to rule out salivary contamination (Citation147). In summary, the EBC apparatus should be designed to maximize condensate collection and minimize the risk of sample contamination.

Many oxidative stress or inflammatory markers have been measured in EBC including H2O2 (Citation42), nitrates (Citation149), leukotrienes (Citation60), among others. However, the potential weaknesses of EBC are the low concentration of detectable biomarkers as compared with induced sputum or bronchoalveolar lavage and the poor reproducibility between studies (Citation151, Citation153). Several recommendations to improve and standardize the collection of EBC are in place, including the use of argon gas to promote biomarker stability and adopting methods to concentrate EBC for easier detection of biomarkers.

CONCLUDING REMARKS

Current research demonstrates a link between oxidative stress and the development of COPD. The oxidative process can be triggered by endogenous factors such as cytokines, pro-inflammatory markers, and immune complexes or by exogenous factors such as pollution and cigarette smoke. Antioxidants may neutralize the induced oxidant burden and have been suggested to play an important role in the prevention of oxidative tissue injury. Recent studies have documented correlations between depleted antioxidant levels and the exacerbation of COPD. Yet, the evidence of different antioxidants and their therapeutic potentials remains limited partly due to the difficulty in the specific sampling of the pulmonary pathway. The exhaled breath analysis thus stands out as a promising tool for the accurate and noninvasive sampling and monitoring of oxidant burden and antioxidant capacity. Ultimately, more research will be needed on EBC of COPD patients to further explore the role of oxidant burden and antioxidant pathways and their potential therapeutic manipulations in the treatment of COPD.

Declaration of interest

The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

REFERENCES

  • Pauling L, Robinson AB, Teranishi R, Cary P. Quantitative analysis or urine vapor and breath by gas-liquid partition chromatography. Proc Natl Acad Sci USA 1971; 68:2374–2376.
  • Lopez AD, Murray CC. The global burden of disease, 1990–2020. Nat Med 1998; 4:1241–1243.
  • Barnes PJ. Immunology of asthma and chronic obstructive pulmonary disease. Nat Rev Immunol 2008; 8:183–192.
  • Joppa P, Petrasova D, Stancak B, Dorkova Z, Tkacova R. Oxidative stress in patients with COPD and pulmonary hypertension. Wien Klin Wochenschr 2007; 119:428–434.
  • Daga MK, Chhabra R, Sharma B, Mishra TK. Effects of exogenous vitamin E supplementation on the levels of oxidants and antioxidants in chronic obstructive pulmonary disease. J Biosci 2003; 28:7–11.
  • Gessner C, Scheibe R, Wotzel M, Hammerschmidt S, Kuhn H, Engelmann L, Hoheisel G, Gillisen A, Sack Ulrich, Wirtz H. Exhaled breath condensate cytokine patterns in chronic obstructive pulmonary disease. Respir Med 2005; 99:1229–1240.
  • Drost EM, Skwarski KM, Sauleda J, Soler N, Roca J, Agusti A, MacNee W. Oxidative stress and airway inflammation in severe exacerbations of COPD. Thorax 2005; 60:293–300.
  • Behrendt CE. Mild and moderate-to-severe COPD in nonsmokers: distinct demographic profiles. Chest 2005; 128:1239–1244.
  • Birring SS, Brightling CE, Bradding P, Entwisle JJ, Vara DD, Grigg J, Wardlaw AJ, Pavord ID. Clinical, radiologic, and induced sputum features of chronic obstructive pulmonary disease in nonsmokers: a descriptive study. Am J Respir Crit Care Med 2002; 166(8):1078–1083.
  • Salvi SS, Barnes PJ. Chronic obstructive pulmonary disease in non-smokers. Lancet 2009; 374(9691):733–743.
  • Schafer FQ, Buettner GR. Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic Biol Med 2001; 30(11):1191–1212.
  • Halliwell, Barry B, Poulsen, Henrik E. Cigarette smoke and oxidative stress. Berlin: Springer, 2006.
  • Hunninghake GW, Crystal RG. Cigarette smoking and lung destruction: accumulation of neutrophils in the lungs of cigarette smokers. Am Rev Respir Dis 1983; 128(5):833–838.
  • van Der Toorn M, Smit-de Vries MP, Slebos DJ, de Bruin HG, Abello N, van Oosterhout AJ, Bischoff R, Kauffman HF. Cigarette smoke irreversibly modifies glutathione in airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 2007; 293(5):L1156–L1162.
  • Foronjy R, Alison W, D’Aarmiento J. The pharmokinetic limitations of antioxidant treatment of COPD. Pulm Pharmacol Ther 2008; 21(2):370–379.
  • Stockley RA. Neutrophils and the pathogenesis of COPD. Chest 2002; 121(5 suppl):151S–155S.
  • Barnes PJ. Alveolar macrophages as orchestrator of COPD. J COPD 2004; 1(1):59–70.
  • Bhowmik A, Seemungal TA, Sapsford RJ, Wedzicha JA. Relation of sputum inflammatory markers to symptoms and lung function changes in COPD exacerbations. Thorax 2000; 55:114–120.
  • Paredi P, Kharitonov SA, Leak D, Ward S, Cramer D, Barnes PJ. Exhaled ethane, a marker of lipid peroxidation, is elevated in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2000; 162:369–373.
  • Tomita K, Barnes PJ, Adcock IM. The effect of oxidative stress on histone acetylation and IL-8 release. Biochem Biophys Res Commun 2003; 301(2):572–577.
  • Barnes PJ. Chronic obstructive pulmonary disease. N Engl J Med 2000; 343(4):269–280.
  • Rahman I. Oxidative stress, chromatin remodeling and gene transcription in inflammation and chronic lung disease. J Biochem Mol Biol 2003; 36(1):95–109.
  • Rajendrasozhan S, Yao H, Rahman I. Current perspective on role of chromatin modifications and deacetylases in lung inflammation in COPD. J COPD 2009; 6(4):291–297.
  • Feldman C, Anderson R, Kanthakumar K, Vargas A, Coles PJ, Wilson R. Oxidant-mediated ciliary dystrunction in human respiratory epithelium. Free Radic Biol Med 1994; 17(1):1–10.
  • Wright DT, Fischer BM, Li C, Rochelle LG, Akley NJ, Adler KB. Oxidant stress stimulates mucin secretion and PLC in airway epithelium via a nitric oxide-dependent mechanism. Am J Physiol 1996; 271(5 pt 1):L854–L861.
  • Kim YD, Jeon JY, Woo HJ, Lee JC, Chung JH, Song SY, Yoon SK, Baek SH. Interleukin-1β induces MUC2 gene expression and mucin secretion via activation of PKC-MEK/ERK, and PL3K in human airway epithelial cells. J Korean Med Sci 2002; 17:765–771.
  • Levine SJ, Larivee P, Logun C, Angus CW, Ognibene FP, Shelhamer JH. Tumor necrosis factor –α induces mucin hyupersecretion and MUC2 gene expression by human airway epithelial cells. Am J Respir Cell Mol Biol 1995; 12:196–204.
  • Yoon JH, Kim KS, Kim HU, Linton JA, Lee JG. Effects of TNF-α and IL-1β on mucin, lysozyme, IL-6 and IL-8 in passage-2 normal human nasal epithelial cells. Acta Otolaryngol 1999; 119:905–910.
  • Li JD, Dohrman AF, Gallup M, Miyata S, Gum JR, Kim YS, Nadel JA, Prince A, Basbaum CB. Transcriptional activation of mucin by pseudomonas aeruginosa lipopolysacchirde in the pathogenesis of cystic fibrosis lung disease. Proc Natl Acad Sci USA 1997; 94:967–972.
  • Demedts IK, Demoor T, Bracke KR, Joos GF, Brusselle GG. Role of apoptosis in the pathogenesis of COPD and pulmonary emphysema. Respir Res 2006; 7:53–62.
  • Anwar S, Whyte MKB. Implications of apoptotic cell death of esident and recruited cells in COPD. J COPD 2008; 5(3):143–145.
  • Segura-Valdez L, Pardo A, Gaxiola M, Uhal BD, Becerril C, Selman M. Upregulation of gelatinases A and B, collagenases 1 and 2, and increased parenchymal cell death in COPD. Chest 2000; 117:684–694.
  • Imai K, Mercer BA, Schulman LL, Sonett JR, D’Armiento JM. Correlation of lung surface area to apoptosis and proliferation in human emphysema. Eur Res J 2005; 25:250–258.
  • Yokohori N, Aoshiba K, Nagai A. Increased levels of cell death and proliferation in alveolar wall cells in patients with pulmonary emphysema. Chest 2004; 125:626–632.
  • Hodge S, Hodge G, Holmes M, Reynolds PN. Apoptosis in COPD. Cur Resp Med Rev 2005; 1:33–41.
  • Park JW, Ryter SW, Choi AMK. Functional significance of apoptosis in chronic obstructive pulmonary disease. J COPD 2007; 4(4):347–353.
  • Fabbri LM, Romagnoli M, Corbetta L, Casoni G, Busljetic K, Turato G, Ligabue G, Ciaccia A, Saetta M, Papi A. Differences in airway inflammation in patients with fixed airflow obstruction due to asthma or chronic obstructive pulmonary disease. Am J Respir Care Med 2003; 167(3):418–424.
  • Di Stefano A, Capelli A, Lusuardi M, Balbo P, Vecchio C, Maestrelli P, Mapp CE, Fabbri LM, Donner CF, Saetta M. Severity of airflow limitation is associated with severity of airway inflammation in smokers. Am J Respir Crit Care Med 1998; 158(4):1277–1285.
  • Bowler RP, Nicks M, Tran K, Tanner G, Cang LY, Young SK, Worthen GS. Extracellular superoxide dismutase attenuates lipopolysaccharide-induced neutrophilic inflammation. Amer J Respir Cell Mol Biol 2004; 31:432–439.
  • Pryor WA, Stone K. Oxidants in cigarette smoke: radicals, hydrogen peroxide, peroxynitrate, peroxynitrite. Annals NY Aca Sci 1993; 686:12–27.
  • Fireman E, Shtark M, Priel IE, Shiner R, Mor R, Kivity S, Fireman Z. Hydrogen peroxide in exhaled breath condensate (EBC) vs eosinophil count in induced sputum (IS) in parenchymal vs airways lung disease. Inflammation 2007; 30:44–51.
  • Chan HP, Tran V, Lewis C, Thomas PS. Elevated levels of oxidative stress markers in exhaled breath condensate. J Thorac Oncol 2009; 4(2):172–178.
  • Vaughan J, Ngamtrakulpanit L, Pajewski TN, Turner R, Nguyen TA, Smith A, Urban P, Hom S, Gaston B, Hunt J. Exhaled breath condensate pH is a robust and reproducible assay of airway acidity. Eur Res J 2003; 22:889–894.
  • Mulkey DK, Henderson RA, Ritucci NA, Putnam RW, Dean JB. Oxidative stress decreases pHi and Na+/H+ exchange and increases excitability of solitary complex neurons from rat brain slices. Am J Physiol Cell Physiol 2004; 286:C940–C951.
  • Kostikas K, Papatheodorou G, Ganas K, Psathakis K, Panagou P, Loukides S. pH in expired breath condensate of patients with inflammatory airway disease. Am J Respir Crit Care Med 2002; 165:1364–1370.
  • Pradico D, Lawson JA, Rokach J, FitzGerald GA. The isoprostanes in biology and medicine. Trends Endocrinol Metab 2001; 12:243–247.
  • Makris D, Paraskakis E, Korakas P, Karagiannakis E, Sourvinos G, Siafakas NM, Tzanakis N. Exhaled breath condensate 8-isoprostane, clinical parameters, radiological indices and airway inflammation in COPD. Respiration 2008; 72(2):138–144.
  • Isik B, Isk SR, Yolacan H, Isik MR. Serum malonyldialdehyde and paraoxonase level in chronic obstructive pulmonary disease. Turk Res J 2005; 6:1.
  • Calikoglu M, Unlu A, Tamer L, Ercan B, Bugdayci R, Atik U. The levels of serum vitamin C, malonyldialdehyde and erythrocyte reduced glutathione in chronic obstructive pulmonary disease and in healthy smokers. Clin Chem Lab Med 2002; 40:10.
  • Balint B, Donnelly E, Hanazawa T, Kharitonov SA, Barnes PJ. Increased nitric oxide metabolites in exhaled breath condensate after exposure to tobacco smoke. Thorax 2001; 56:456–461.
  • Kharitonov SA, Barnes PJ. Nitric oxide, nitrotyrosine, and nitric oxide modulators in asthma and chronic obstructive pulmonary disease. Curr Allergy Asthma Rep 2003; 3:121–129.
  • Osoata G, Hanazawa T, Brindicci C, Ito M, Barnes PJ, Kharitonov S, Ito K. Peroxynitrite elevation in exhaled breath condensate of COPD and its inhibition by fudosteine. Chest 2009; 135:1513–1520.
  • Montuschi P, Kharitonov SA, Barnes PJ. Exhaled carbon monoxide and nitric oxide in COPD. Chest 2001; 120(2):496–501.
  • Brindicci C, Ito K, Resta O, Pride NB, Barnes PJ, Kharitono SA. Exhaled nitric oxide from lung periphery is increased in COPD. Eur Respir J 2005; 26:52–59.
  • Ansarin K, Chatkin JM, Ferreira IM, Gutierrez CA, Zamel N, Chapman KR. Exhaled nitric oxide in chronic obstructive pulmonary disease: relationship to pulmonary function. Eur Respir J 2001; 17:934–938.
  • Ji Y, Neverova I, Van Eyk JE, Bennett BM. Nitration of tyrosine 92 mediates the activation of rat microsomal glutathione S-transferase by peroxynitrite. J Biol Chem 2006; 281:1986–1991.
  • Balint B, Kharitonov SA, Hanazawa T, Donnelly LE, Shah PL, Hodson ME, Barnes PJ. Increased introtyrosine in exhaled breath condensate in cystic fibrosis. Eur Res J 2001; 17:1201–1207.
  • Ueshima K, Minakata Y, Sugiura H, Yanagisawa S, Ichikawa T, Akamatsu K, Hirano T, Nakanishi M, Matsunaga K, Yamagata T, Ichinose M. The influence of free 3-nitrotyrosine and saliva on the quantitative analysis of protein-bound 3-nitrotyrosine in sputum. Anal Chem Insights 2007; 2:1–7.
  • 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. Nitrosative stress in the bronchial mucosa of severe chronic obstructive pulmonary disease. J Allergy Clin Immunol 2005; 116(5):1028–1035.
  • Wung BS, Cheng JJ, Hsieh HJ, Shyy YJ, Wang DL. Cyclic strain-induced monocyte chemotactic protein-1 gene expression in endothelial cells involves reactive oxygen species activation of activator protein 1. Circ Res 1997; 8:1–7.
  • Carpagnano GE, Kharitonov SA, Foschino-Barbaro MP, Resta O, Garmiccioni E, Barnes PJ. Increased inflammatory markers in the exhaled breath condensate of cigarette smokers. Eur Res J 2003; 21:589–593.
  • Montuschi P, Kharitonov S, Ciabattoni G, Barnes PJ. Exhaled leukotrienes and prostaglandins in COPD. Thorax 2003; 58(7):585–588.
  • Scheideler L, Manke HG, Schwulera U, Inacker O, Hammerle H. Detection of nonvolatile macromolecules in breath: a possible diagonstic tool? Am Rev Respir Dis 1993; 148:778–784.
  • Cunningham S, McColm JR, Ho LP, Greening AP, Marshall TG. Measurement of inflammatory markers in the breath condensate of children with cystic fibrosis. Eur Res J 2000; 15:955–957.
  • Cao WQ, Duan YX. Breath analysis: potential for clinical diagnosis and exposure assessment. Clin Chem 2006; 52(5):800–811.
  • Rovina N, Papapetropoulos A, Kollintza A, Michailidou M, Simoes D, Roussos C, Gratziou C. Vascular endothelial growth factor: an angiogenic factor reflecting airway inflammation in healthy smokers and in patients with bronchitis type of chornic obstructive pulmonary disease. Respir Res 2007; 8:53–60.
  • Garey KW, Neuhauser MM, Robbins RA, Danziger LH, Rubinstein I. Markers of inflammation in exhaled breath condensate of young healthy smokers. Chest 2004; 125(1):22–25.
  • Sapey E, Bayley D, Ahmad A, Newbold P, Snell N, Stockley RA. Inter-relationships between inflammatory markers in patients with stable COPD with bronchitis: intra-patient and inter-patient variability. Thorax 2007; 63:493–499.
  • Moore B, Murphy RF, Agrawal DK. Interaction of TGF-beta with immune cells in airways disease. Curr Mol Med 2008; 8(5):427–436.
  • Duvernelle C, Freund V, Frossard N. Transforming growth factor-beta and its role in asthma. Pulm Pharmacol Ther 2003; 16(4):181–196.
  • Van Berkel JJBN, Dallinga JW, Moller GM, Godschalk RWL, Moonen EJ, Wouters EFM, Van Schooten FJ. A profile of volatile organic compounds in breath discriminates COPD patients from controls. Respir Med 2009; 104(4):557–563.
  • Ibrahim B, Basanta M, Dockry R, Tal-Singer R, Douce D, Woodcock AA, Singh D, Fowler SJ. Exhaled volatile organic compounds as potential biomarkers in chronic obstructive pulmonary disease. Amer J Respir Crit Care Med 2010; 181:A1012.
  • Kanazawa H, Yoshikawa J. Elevated oxidative stress and reciprocal reduction of vascular endothelial growth factor levels with severity of COPD. Chest 2005; 128:3191–3197.
  • Kasahara Y, Tuder RM, Cool CD, Lynch DA, Flores SC, Voelkel NF. 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; 163:737–744.
  • Sakao S, Tatsumi K, Hashimoto T, Igari H, Shino Y, Shirasawa H, Kuriyama T. Vascular endothelial growth actor and the risk of smoking-related COPD. Chest 2003; 124:323–327.
  • Day BJ. Antioxidants as potential therapeutics for lung fibrosis. Antioxid Redox Signal 2008; 10(2):355–370.
  • MacNee W. Treatment of stable COPD: antioxidants. Eur Respir Rev 2005; 14:12–22.
  • Oberley-Deegan RE, Regan EA, Kinnula VL, Crapo JD. Extracellular superoxide dismutase and risk of COPD. J COPD 2009; 6(4):307–312.
  • Larsen GL, White CW, Takeda K, Loader JE, Nguyen DD, Joetham A, Groner Y, Gelfand EW. Mice that overexpress Cu/Zn superoxide dismutase are resistant to allergen-induced changes in airway control. Am J Physiol Lung Cell Mol Physiol 2000; 279(2):L350–L359.
  • White CW, Avraham KB, Shanley PF, Groner Y. Transgenic mice with expression of elevated levels of copper–zinc superoxide dismutase in the lungs are resistant to pulmonary oxygen toxicity. J Clin Invest 1991; 87(6):2162–2168.
  • Chang LY, Kang BH, Slot JW, Vincent R, Crapo JD. Immunocytochemical localization of the sites of superoxide dismutase induction by hyperoxia in rat lungs. Lab Invest 1995; 73(1):29–39.
  • Wispe JR, Warner BB, Clark JC, Dey CR, Neuman J, Glasser SW, Crapo JD, Chang LY, Whitsett JA. Human Mn-superoxide dismutase in pulmonary epithelial cells of transgenic mice confers protection from oxygen injury. J Biol Chem 1992; 267(33):22937–22941.
  • Harju T, Kaarteenaho-Wiik R, Sirvio R, Paakko P, Crapo JD, Oury TD, Shoini Y, Kinnula VL. Manganese superoxide dismutase is increased in the airways of smokers lungs. Eur Res J 2004; 24:765–771.
  • Oury TD, Chang LY, Marklund SL, Day BJ, Crapo JD. Immunocytochemical localization of extracellular superoxide dismutase in human lung. Lab Invest J Tech Meth Pathol 1994; 70:889–898.
  • Oury TD, Day BJ, Crapo JD. Extracellular superoxide dismutase in vessels and airways of humans and baboons. Free Radic Biol Med 1996; 20:957–965.
  • Gao F, Kinnula VL, Myllarniemi M, Oury TD. Extracellular superoxide dismutase in pulmonary fibrosis. Antioxid Redox Signal 2008; 10:343–354.
  • Reinhard C, Meyer B, Fuchs H, Stoeger T, Eder G, Ruschendorf F, Heyder J, Nurnberg P, De Angelis MH, Schulz H. Genomewide linkage analysis identifies novel genetic loci for lung function in mice. Am J Respir Crit Care Med 2005; 171:880–888.
  • Ganguly K, Stoeger T, Wesselkamper SC, Reinhard C, Sartor MA, Medvedovic M, Tomlinson CR, Bolle I, Mason JM, Leikauf GD, Schulz H. Candidate genes controlling pulmonary function in mice: transcript profiling and predicted protein structure. Physiol Genom 2007; 31:410–421.
  • Wilk JB, Walter RE, Laramie JM, Gottlieb DJ, O’Connor GT. Framingham Heart Study genome-wide associations: results for pulmonary function measures. BMC Med Genetics 2007; 8(Supp 1): S8.
  • Young RP, Hopkins R, Black PN, Eddy C, Wu L, Gamble GD, Mills GD, Garret JE, Eaton TE, Rees MI. Functional variants of antioxidant genes in smokers with COPD and in those with normal lung function. Thorax 2006; 61:394–399.
  • 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.
  • Harju T, Mazur W, Merikallio H, Soini Y, Kinnula VL. Glutathione-S-transferases in lung and sputum specimens, effects of smoking and COPD serverity. Respiratory Research 2008, 9:80, doi:10.1186/1465-9921-9-80.
  • Joshi UM, Kodavanti PR, Mehendale HM. Glutathione metabolism and utilization of external thiols by cigarette smoke-challenge, isolated rat and rabbit lungs. Toxicol Appl Pharmacol 1988; 96:324–335.
  • Park EM, Park YM, Gwak YS. Oxidative damage in tissues of rats exposed to cigarette smoke. Free Radic Biol Med 1998; 25:79–86.
  • Cantin AM, North SL, Hubbard RC, Crystal RG. Normal alveolar epithelial lining fluid contains high levels of glutathione. J Appl Physiol 1987; 63:152–157.
  • Wang Y, Feinstein SI, Fisher AB. Peroxiredoxin 6 as an antioxidant enzyme protection of lung alveolar epithelial type II cells from HO-induced oxidative stress. J Cell Biochem 2008; 104:1274–1285.
  • Pak JH, Manevich Y, Kim HS, Feinstein SI, Fisher AB. An antisense oligonucleotide to 1-cys peroxiredoxin causes lipid peroxidation and apoptosis in lung epithelial cells. J Biol Chem 2002; 277:47729–49934.
  • Wang Y, Manevich Y, Feinstein SI, Fisher AB. Adenovirus-mediated transfer of the 1-cys peroxiredoxin gene to mouse lung protects against hyperoxic injury. Am J Physiol Lung Cell Mol Physiol 2004; 286:L1188–L1193.
  • Wang Y, Phelan SA, Manevich Y, Feinstein SI, Fisher AB. Transgenic mice overexpressing peroxiredoxin 6 show increased resistance to lung injury in hyperoxia. Am J Respir Cell Mol Biol 2006; 34:481–486.
  • Rizvi SI, Maurya PK. Alterations in antioidant enzymes during aging in humans. Mol Biotechnol 2007; 37(1):58–61.
  • Moukdar F, Robidoux J, Lyght O, Pi J, Daniel KW, Collins S. Reduced antioxidant capacity and diet-induced atherosclerosis in uncoupling protein-2-deficient mice. J Lipid Res 2009; 50:59–70.
  • Inomata T, Betsuyaku T, Ito Y, Nasuhara Y, Nishimura M. Catalse expression in pulmonary macrophages of smokers and chronic obstructive pulmonary disease (COPD) patients. Amer J Respir Crit Care Med 2009; 179:A4157.
  • Mak JCW, Ho SP, Yu WC, Choo KL, Chu CM, Yew WW, Lam WK, Chan-Yeung M. Polymorphisms and functional activity in superoxide dismutase and catalase genes in smokers with COPD. Eur Respir J 2007; 30:684–690.
  • Stocker R, Yamamoto Y, McDonagh AF, Glazer AN, Ames BN. Bilirubin is an antioxidant of possible physiological importance. Science 1987; 235:1043–1046.
  • Shinohara T, Kaneko T, Nagashima Y, Ueda A, Tagawa A, Ishigatsubo Y. Adenovirus-mediated transfer and overexpression of heme oxygenase 1 cNDNA in lungs attenuates elastase-induced pulmonary emphysema in mice. Hum Gene Ther 2005; 16:318–327.
  • Fredenburgh LE, Perrella MA, Mitsialis SA. The role of heme oxygenase-1 in pulmonary disease. Am J Respir Cell Mol Biol 2007; 36:158–165.
  • Choi BM, Pae HO, Jeong YR, Oh GS, Jun CD, Kim BR, Kim YM, Chung HT. Overexpression of heme oxygenase (HO)-1 renders Jurkat T cells resistant to fas-mediated apoptosis: involvement of iron released by HO-1. Free Radic Biol Med 2004; 36:858–871.
  • Sarady JK, Otterbein SL, Liu F, Otterbein LE, Choi AM. Carbon monoxidemodulates endotoxin-induced production of granulocyte macrophage colony-stimulating factor in macrophages. Am J Respir Cell Mol Biol 2002; 27:739–745.
  • Guenegou A, Leynaert B, Pin I, Le Moel G, Zureik M, Neukirch F. Serum carotenoids, vitamins A and E, 8 year lung function decline in a general population. Thorax 2006; 61(4):320–326.
  • Wu TC, Huang YC, Hsu SY, Wang YC, Yeh SL. Vitamin E and vitamin C supplementation in patients with chronic obstructive pulmonary disease. Int J Vitam Nutr Res 2007; 77(4):272–279.
  • Cotgreave IA. N-acetylcysteine: pharmacological considerations and experimental and clinical applications. Adv Pharmacol 1997; 38:205–227.
  • Stav D, Raz M. Effect of N-acetylcysteine on airtrapping in COPD: a randomized placebo-controlled study. Chest 2009; 136(2):381–386.
  • Committee BTSR. Oral N-acetylcysteine and exacerbation rates in patients with chronic bronchitis and severe airways obstruction. Thorax 1985; 40(11):832–835.
  • Grassi C, Morandini GC. A controlled trial of intermittent oral acetylcysteine in the long-term treatment of chronic bronchitis. Eur J Clin Pharmacol 1976; 9(5–6):393–396.
  • Hansen NC, Skriver A, Brorsen-Riis L, Balslov S, Evald T, Maltbaek N, Gunnersen G, Garsdal P, Sander P, Pedersen JZ, Ibsen TB, Rasmussen FV. Orally administered N-acetylcysteine may improve general well-being in patietns with mild chronic bronchitis. Respir Med 1994; 88(7):531–535.
  • Schermer T, Chavannes N, Dekhuijzen R, Wouters E, Muris J, Akkermans R, van Schayck O, van Weel C. Fluticasone and N-acetylcysteine in primary care patietns with COPD or chronic bronchitis. Respir Med 2009; 103(4):542–551.
  • Decramer M, Rutten-van Molken M, Dekhuijzen PN, Troosters T, van Herwaarden C, Pellegrino R, van Schayck CP, Olivieri D, Del Donno M, De Backer W, Lankhorst I, Ardia A. Effects of N-acetylcysteine on outcomes in chronic obstructive pulmonary disease (Bronchitis Randmoized on NAC Cost-Utility Study, BRONCUS): a randomized placebo-controlled trial. Lancet 2005; 365:1552–1560.
  • Schwartz J, Weiss ST. Dietary factors and their relation to respiratory symptoms. The second National Health and Nutrition Examination Survey. Am J Epidemiol 1990; 132:67–76.
  • Schwartz J, Weiss ST. Relationship between dietary vitamin C intake and pulmonary function in the first national health and nutrition examination survey (NHANES I). Am J Clin Nutr 1994; 59:110–114.
  • Hu G, Cassano PA. Antioxidant nutrients and pulmonary function: the Third National Health and Nutrition Examination Survey (NHANES III). Am J Epidemiol 2000; 151:975–981.
  • Tabak C, Feskens EJ, Heederik D, Kromhout D, Menotti A, Blackburn HW. Fruit and fish consumption: a possible explanation for population differences in COPD mortality (The Seven Countries Study). Eur J Clin Nutr 1998; 52:819–825.
  • Tabak C, Smit HA, Heederik D, Ocke MC, Kromhout D. Diet and chronic obstructive pulmonary disease: inderdependent beneficial effects of fruits, whole grains and alcohol (the MORGAN Study). Clin Exp Allergy 2001; 31:747–755.
  • Miedema I, Feskens EJ, Heederik D, Kromhout D. Dietary determinants of long-term incidence of chronic non-specific lung disease: The Zutphen Study. Am J Epidemiol 1993; 138:37–45.
  • Gilks CB, Price K, Wright JL, Churg A. Antioxidant gene expression in rat lung after exposure to cigarette smoke. Am J Pathology 1998; 152:269–278.
  • Tager M, Piecyk A, Kohnlein T, Thiel U, Ansorge S, Welte T. Evidence of a defective thiol status of alveolar macrophages from COPD patients and smokers: chronic obstructive pulmonary disease. Free Radic Biol Med 2000; 29(11):1160–1165.
  • Rahman I, Morrison D, Donaldson K, MacNee W. Systemic oxidative stress in asthma, COPD and smokers. Am J Respir Crit Care Med 1996; 154(4 pt 1):1055–1060.
  • Celli BR, Barnes PJ. Exacerbations of chronic obstructive pulmonary disease. Eur Respir J 2007; 29:1224–1238.
  • Varkey JB, Varkey B. Viral infections in patients with chronic obstrutive pulmonary disease. Curr Opin Pulm Med 2008; 14:89–94.
  • Novel Swine-Origin Influenza A (H1N1) Virus Investiation Team. Emergence of a novel swine-origin influenza A (H1N1) virus in humans. N Engl J Med 2009; 360:2605–2615.
  • Hansel TT, Barnes PJ. New drugs for exacerbations of chronic obstructive pulmonary disease. Lancet 2009; 374:744–755.
  • Biernacki WA, Kharitonov SA, Barnes PJ. Increased leukotriene B4 and 8-isoprostane in exhaled breath condensate of patients with exacerbation of COPD. Thorax 2003; 58(4):294–298.
  • Drost EM, Ritchie HE, Skwarska E. NF-κB activation in airway leucocytes in COPD patients. Eur Res J 2001; 160:260s.
  • Nishikawa M, Kakemizu N, Ito T, Kudo M, Kaneko T, Suzuki M, Udaka N, Ikeda H, Okubo T. Superoxide mediates cigarette smoke-induced infiltration of neutrophils into the airways through nuclear factor-kappaB activation and IL-8 mRNA expression in guinea pigs in vivo. Am J Respir Cell Mol Biol 1999; 20:189–198.
  • Tuder RM, Zhen L, Cho CY, Taraseviciene-Stewart L, Kasahara Y, Salvemini D, Voelkel NF, Flores SC. Oxidative stress and apoptosis interact and cause emphysema due to vascular endothelial growth factor receptor blockade. Am J Respir Cell Mol Biol 2003; 29:88–97.
  • Hirayama F, Lee AH, Binns CW, Zhao Y, Hiramatsu T, Tanikawa Y, Nishimura K, Taniguchi H. Do vegetables and fruits reduce the risk of chronic obstructive pulmonary disease? A case–control study in Japan. Prev Med 2009; 49(2–3):184–189.
  • Sutherland ER, Crapo JD, Bowler RP. N-acetylcysteine and exacerbations of chronic obstructive pulmonary disease. J COPD 2006; 3(4):195–202.
  • Zuin R, Palamidese A, Negrin R, Catozzo L, Scarda A, Balbinot M. High-dose N-acetylcysteine in patietns with exacerbations of chronic obstructive pulmonary disease. Clin Drug Investig 2005; 25(6):401–408.
  • Epperly MW, Defilippi S, Sikora C, Gretton J, Kalend A, Greenberger JS. Intractracheal injection of manganese superoxide dismutase (MnSOD) plasmid/liposomes protects normal lung but not orthotopic tumors from irradiation. Gene Therapy 2000; 7:1011–1018.
  • Griese M, Ramakers J, Krasselt A, Starosta V, Van Koningsbruggen S, Fischer R, Ratjen F, Müllinger B, Huber RM, Maier K, Rietschel E, Scheuch G. Improvement of alveolar glutathione and lung function but not oxidative state in cystic fibrosis. Am J Respir Crit Care Med 2004; 169:822–828.
  • Bishop C, Hudson VM, Hilton SC, Wild C. A pilot study of the effect of inhaled buffered glutathione on the clinical status of patients with cystic fibrosis. Chest 2005; 127:308–317.
  • MacNee W, Rahman I. Oxidants/antioxidants in idiopathic pulmonary fibrosis. Thorax 1995; 50(Supp 1):S53–S58.
  • Borok Z, Buhl R, Grimes GJ, Bokser AD, Hubbard RC, Holroyd KJ, Roum JH, Czerski DB, Cantin AM, Crystal RG. Effect of glutathione aerosol on oxidant-antioxidant imbalance in idiopathic pulmonary fibrosis. Lancet 1991; 338(8761):215–216.
  • Gillissen A, Jaworska M, Orth M, Coffiner M, Maes P, App EM, Cantin AM, Schultze-Werninghaus G. Nacystelyn, a novel lysine salt of N-acetylcysteine, to augment cellular antioxidant defence in vitro. Respir Med 1997; 91:159–168.
  • Mao JT, Tashkin DP, Belloni PN, Baileyhealy I, Baratelli F, Roth MD. All-trans retinoic acid modulates the balance of matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 in patients with emphysema. Chest 2003; 125(4):1724–1732.
  • Calverley PM, Sanchez-Toril F, McIvor A, Teichmann P, Bredenbroeker D, Fabbri LM. Effect of 1-year treatment with roflumilast in severe chronic obstructive pulmonary disease. Am J Respir Care Med 2007; 176(2):154–161.
  • Vestbo J, Tan L, Atkinson G, Ward J. and on behalf of the UK-500,001 Global Study Team. A controlled trial of 6-weeks’ treatment with a novel inhaled phophodiesterase type-4 inhibitor in COPD. Eur Respir J 2009; 33(5):1039–1044.
  • Mutlu GM, Garey KW, Robbins RA, Danziger LH, Rubinstein I. Collection and analysis of exhaled breath condensate in humans. Am J Respir Crit Care Med 2001; 164:731–737.
  • Montuschi P. Review: analysis of exhaled breath condensate in respiratory medicine: methodological aspects and potential clinical applications. Ther Adv Res Dis 2007; 1:5–23.
  • Liu J, Thomas PS. Exhaled breath condensate as a method of sampling airway nitric oxide and other markers of inflammation. Med Sci Monit 2005; 11(8):MT53–MT62.
  • Kharitonov SA, Barnes PJ. Exhaled Biomarkers. Chest 2006; 131:1541–1546.
  • Liu J, Conrad DH, Chow S, Tran VH, Yates DH, Thomas PS. Collection devices influence the constituents of exhaled breath condensate. Eur Respir J 2007; 30:807–808.
  • Silkoff PE, Erzurum SC, Lundberg JO, George SC, Marczin N, Hunt JF, Effros R, Horvath I. ATS Workshop Proceedings: exhaled nitric oxide and nitric oxide oxidative metabolism in exhaled breath condensate. Proc Am Thorac Soc 2006; 3(2):131–145.
  • Chow S, Yates DH, Thomas PS. Reproducibility of exhaled breath condensate markers. Eur Respir J 2008; 32(4):1124–1126.
  • Lee W, Thomas PS. Oxidative stress in COPD and its measurement through exhaled breath condensate. Clin Transl Sci 2009; 2(2):150–155.
  • Dahl M, Bowler RP, Juul K, Crapo JD, Levy S, Nordestgaard BG. Superoxide dismutase 3 polymorphism associated with reduced lung function in two large population. Amer J Respir Crit Care Med 2008; 178(9):906–912.
  • Arcaroli JJ, Hokanson JE, Abraham E, Geraci M, Murphy JR, Bowler RP, Dinarello CA, Silveira L, Sankoff J, Heyland D, Wischmeyer P, Crapo JD. Extracellular superoxide dismutase haplotypes are associated with acute lung injury and mortality. Amer J Respir Crit Care Med 2009; 179:105–112.
  • Sagiura H, Ichikawa T, Liu X, Kobayashi T, Wang XQ, Kawasaki S, Togo S, Kamio K, Mao L, Ann Y, Ichinose M, Rennard SI. N-acetyl-l-cysteine inhibits TGF-beta(1)-induced profibrotic response in fibroblasts. Pulm Pharmacol Ther 2009; 22(6):487–491.
  • Decramer M, Dekhuijzen PNR, Troosters T, Van Herwaarden C, Rutten-Van Molken M, Van Schayck CPO, Olivieri D, Lankhorst I, Ardia A. The Bronchitis Randomized on NAC Cost-Utility Study (BRONCUS): hypothesis and design. Eur Res J 2001; 17:329–336.
  • Kirkil G, Hamdi Muz M, Seckin D, Sahin K, Kucuk O. Antioxidant effect of zinc picolinate in patietns with chronic obstructive pulmonary disease. Respir Med 2008; 102(6):840–844.
  • Nadeem A, Raj HG, Chhabra SK. Effect of vitamin E supplementation with standard treatment on oxidant–antioxidant status in chronic obstructive pulmonary disease. Indian J Med Res 2008; 128(6):705–711.
  • Conklin JD, Trinity JD, Amann M, Fjeldstad A, Wray DW, Richardson RS. Antioxidant supplementation improves oxygen delivery and blood flow to exercising skeletal muscle in COPD. Amer J Respir Crit Care Med 2010; 181:A6842.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.