Next Article in Journal
Residue and Risk Assessment of Fluopyram in Carrot Tissues
Previous Article in Journal
Scoping Review: Evaluation of Moringa oleifera (Lam.) for Potential Wound Healing in In Vivo Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential of Small Molecules Targeting Oxidative Stress in the Treatment of Chronic Obstructive Pulmonary Disease (COPD): A Comprehensive Review

by
Hamad Ghaleb Dailah
Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan 45142, Saudi Arabia
Molecules 2022, 27(17), 5542; https://doi.org/10.3390/molecules27175542
Submission received: 25 July 2022 / Revised: 21 August 2022 / Accepted: 25 August 2022 / Published: 28 August 2022

Abstract

:
Chronic obstructive pulmonary disease (COPD) is an increasing and major global health problem. COPD is also the third leading cause of death worldwide. Oxidative stress (OS) takes place when various reactive species and free radicals swamp the availability of antioxidants. Reactive nitrogen species, reactive oxygen species (ROS), and their counterpart antioxidants are important for host defense and physiological signaling pathways, and the development and progression of inflammation. During the disturbance of their normal steady states, imbalances between antioxidants and oxidants might induce pathological mechanisms that can further result in many non-respiratory and respiratory diseases including COPD. ROS might be either endogenously produced in response to various infectious pathogens including fungi, viruses, or bacteria, or exogenously generated from several inhaled particulate or gaseous agents including some occupational dust, cigarette smoke (CS), and air pollutants. Therefore, targeting systemic and local OS with therapeutic agents such as small molecules that can increase endogenous antioxidants or regulate the redox/antioxidants system can be an effective approach in treating COPD. Various thiol-based antioxidants including fudosteine, erdosteine, carbocysteine, and N-acetyl-L-cysteine have the capacity to increase thiol content in the lungs. Many synthetic molecules including inhibitors/blockers of protein carbonylation and lipid peroxidation, catalytic antioxidants including superoxide dismutase mimetics, and spin trapping agents can effectively modulate CS-induced OS and its resulting cellular alterations. Several clinical and pre-clinical studies have demonstrated that these antioxidants have the capacity to decrease OS and affect the expressions of several pro-inflammatory genes and genes that are involved with redox and glutathione biosynthesis. In this article, we have summarized the role of OS in COPD pathogenesis. Furthermore, we have particularly focused on the therapeutic potential of numerous chemicals, particularly antioxidants in the treatment of COPD.

1. Introduction

Chronic obstructive pulmonary disease (COPD) is a chronic respiratory disease. One of the very common characteristics of COPD includes irreversible obstructive breathing. The World Health Organization (WHO) identified COPD as the third leading cause of death worldwide in 2019 [1]. Oxidative stress (OS) has significant effects on various lung functions and in COPD pathogenesis. These effects include apoptosis, remodeling of the extracellular matrix, alveolar epithelial injury, mitochondrial respiration, membrane lipid peroxidation (LPO), mucus hypersecretion, and oxidative inactivation of surfactants and antiproteases [2,3]. In the respiratory tracts of COPD patients, an elevated level of OS is observed because of the elevated oxidant burden from environmental exposure including air pollutants and cigarette smoke (CS), and from the elevated levels of reactive nitrogen species (RNS) and reactive oxygen species (ROS) secreted from the macrophages and leukocytes associated with the inflammatory processes in the lungs of COPD patients [4,5,6]. These RNS and ROS have the capacity to cause oxidative damage to proteins, carbohydrates, lipids, and DNA, which can further lead to various downstream mechanisms that can lead to COPD development and progression. In addition, they can also cause the activation of resident cells in the lung including alveolar macrophages and epithelial cells to produce chemotactic molecules that engage more inflammatory cells (such as lymphocytes, monocytes, and neutrophils) into the lung [4,7,8], which eventually result in the spread of OS in the lung. As a whole, these processes result in a dangerous cycle of tenacious inflammation along with chronic OS, which can result in faulty tissue repair processes, disruptions in the protease–antiprotease balance, increased autophagy, and apoptosis, which can eventually play roles in the progression and severity of COPD [9,10,11,12].
ROS is the main cause of tissue and cell injury linked with numerous inflammatory pulmonary diseases including COPD [13,14]. Nonetheless, the exact mechanisms of COPD pathogenesis are yet to be fully revealed. Oxidant/antioxidant imbalance and elevated levels of OS may be the main causes of COPD. An elevated level of OS is produced from airway leukocytes in the blood or in air spaces indirectly because of the secretion of elevated levels of ROS, and directly because of the environmental oxidant pollutants and CS [15]. It is well-known that antioxidant enzymes and compounds scavenge ROS [13,16]. Cigarette smoking is the major etiological factor in COPD pathogenesis, since it can result in OS in the lower airways. Furthermore, CS contains over 1016–1017 oxidants per puff and around 4700 chemicals such as nitrogen oxides, superoxide radicals, and peroxynitrite (ONOO) [17]. CS exerts significant adverse effects; nonetheless the effects of other risk factors also need to be well-considered, since not all smokers develop COPD. In Table 1, we summarized the risk factors that are linked with COPD development. Multiple intracellular and extracellular antioxidants provide protection to the blood and lungs against the harmful activities of oxidants, under normal conditions [15]. It has been reported that antioxidants including thiol compounds/donors and their analogs including glutathione (GSH) and mucolytic drugs, for example, fudosteine, erdosteine, carbocysteine, and N-acetyl-L-cysteine have the ability to efficiently regulate nuclear factor-κB (NF-κB) activation, elevate intracellular thiol concentrations, and detoxify/scavenge oxidants/radicals, therefore suppressing inflammatory gene expressions [18]. In this article, we have highlighted the important role of OS in COPD pathogenesis. We have also particularly focused on the therapeutic potential of numerous antioxidants in the treatment of COPD.

2. Reactive Oxygen Species

OS is supposed to be triggered via CS, and OS-mediated cell injury has a significant contribution in the development of COPD [47]. CS contains a complex combination of many free radicals and ROS, which is further grouped into two phases including tar and gas. The tar phase contains around 1017 long-lived free radicals per gram, for instance, quinone/hydroquinone (Q/QH2) radicals can decrease oxygen to generate superoxide anions (O2•−), resulting in the production of hydroxyl radicals (OH) (Table 2) and hydrogen peroxide (H2O2) [48,49]. It has been reported that OH is a highly reactive ROS, which can injure nearly all types of macromolecules following collision [50]. Interestingly, hydroxyl radicals can be produced via a Fenton reaction including H2O2 and cuprous copper (Cu(I)) or ferrous iron (Fe(II)), which comprise harmful connections of redox- and metal homeostasis. Particulate matter (also known as particle pollution) increases OS, which can further damage the Fenton reaction and lead to the generation of OH in the lung [51]. Superoxide radical anions (O2•−) are less reactive compared to OH, however, O2•− is also detrimental and can play a role in the one-electron pathway involving flavin cofactors and metals. In contrast, H2O2 is comparatively stable, which has the capacity to travel long distances from its location of generation [50].
Compared to the superoxide radical anions, H2O2 mainly participates in two-electron pathways involving sulfur-containing moieties in the cell. Nonetheless, H2O2 also plays a role in some one-electron pathways involving transition metals. Interestingly, H2O2 can play a role as a damaging agent at higher concentrations and as a signaling molecule at low concentrations. Therefore, H2O2 exerts a significant cellular effect that is defined via overlapping processes of H2O2 identification, signal transduction, and obliteration [52,53]. Hypochlorous acid (HOCl) produced in the presence of H2O2 can result in the generation of more harmful ROS including OH [54]. Hypochlorite anions (OCl) exhibit increased reactivity, which suggests that it indiscriminately modifies its targets, usually with second-order rate constants of 105–107  M−1 s−1 [55]. In the case of proteins, various amino acids including methionine, histidine, and cysteine are known as the ideal residues for modification. Moreover, OCl has the capacity to alter various primary amines (those are present in the lysine’s sidechain) to chloramines. Collectively, increased concentrations of ROS might result in respiratory problems and lung tissue injury by modifying several target molecules by different ROS-specific processes.
On the other hand, the gas phase of CS possesses an increased level of reactive molecules compared to tar. In addition, the gas phase possesses 1015 inorganic and organic radicals per puff including ONOO, nitrogen dioxide, and nitric oxide (NO) [49,56]. NO is one of the major RNS and ROS. CS also possesses around 74.5–1008 ppm NO and smokers are mostly exposed to this free radical. Although NO has a shorter half-life (t1/2= around 0.09–2 s), this free radical reacts rapidly with O2•− to generate ONOO− [49,57,58,59]. ONOO (another RNS) is associated with numerous pathological and physiological mechanisms [60,61]. ONOO also has powerful nitration and oxidation capacities, which can further damage various molecules in the cells including proteins and DNA. Interestingly, a second-order reaction relies on the levels of two first-order reactants or one second-order reactant, which are NO and O2•− in the generation of ONOO. NO also has the capacity to interact with organic lipid peroxyl radicals (ROO) (that are found in CS) to generate cytotoxic species including alkyl peroxynitrites (ROONO). It has been observed that O2•− and NO are generated via multiple inflammatory cells including macrophages via nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complexes (NOXs) and nitric oxide synthases (NOSs), respectively. In the endoplasmic reticulum, RNS and ROS can also be secreted via an uncontrolled mechanism as by-products during various processes including the protein folding maturation process, peroxisomal metabolism, and mitochondrial respiration [62,63]. Moreover, their elevated generation can eventually result in OS and lung injury.
Table 2. A summary of the free radicals that can play a role in oxidative stress.
Table 2. A summary of the free radicals that can play a role in oxidative stress.
NameStructural FormulaPropertiesReferences
Reactive oxygen species
Radicals
Hydroxyl radicalOHHighly reactive, very unstable in aqueous solutions [64]
SuperoxideO2Moderately reactive, highly unstable, modulate signaling[65,66]
Peroxyl radicalROOProducts of lipid peroxidation[67]
Alkoxyl radicalROProducts of lipid peroxidation[67]
Non Radicals
Hydrogen peroxideH2O2Toxic, associated with several signal transduction pathways and cell fate decisions[68,69]
Hypochlorite anionOClProduced by myeloperoxidase[70]
Singlet oxygen1O2highly excited, nonradical, metastable state of molecular oxygen [67]
OzoneO3Environmental air pollutant[71]
Reactive nitrogen species
Radicals
Nitrogen dioxideNO2One of the most threatening environmental air pollutants, highly reactive[65]
Nitric oxideNOImportant redox signaling molecule[72]
Non Radicals
Nitrogen oxidesNOxEnvironmental toxins including NO and NO2 linked with combustion sources[65,73]
PeroxynitriteONOOHighly reactive, unstable intermediate[74,75]

3. Endogenous and Exogenous Generations of Reactive Oxygen Species

The lungs can be exposed to ROS and RNS derived from environmental and cellular sources. CS is the major environmentally derived ROS that induces COPD pathogenesis. RNS/ROS can also be produced via multiple structural and inflammatory cells of the airways. The inflammatory response (inflammation) is a feature of COPD, which is characterized via activation of resident macrophages and epithelial cells, and the recruitment and activation of monocytes, neutrophils, and T- and B-lymphocytes. Once employed in the airspace, inflammatory cells become activated and produce ROS as a reaction to an adequate concentration of a secretagogue stimulus (threshold condition). In inflammatory cells, NADPH oxidase (NOX) is the major ROS-generating enzyme. Various other enzyme systems including the heme peroxidases and xanthine/xanthine oxidase (XO) system are also associated with COPD [76,77]. In a similar manner, nitric oxide synthase mediates the generation of RNS in the form of nitric oxide (NO) generation. In the presence of superoxide anions, nitric oxide generates more strong and harmful ONOO molecules. It has been reported that the disturbance of the components of NOX including gp91phox and p47phox, which was found to enlarge the airspace in mouse models, further indicating that ROS-derived from NOX can play a role in the signaling pathways of tissue homeostasis. Moreover, in COPD, the use of inhibitors of NOX to redress the imbalance between antioxidants and oxidants might be harmful [78].

4. Sources of Reactive Oxygen Species in the Lung

The lung is susceptible to damage from environmental OS due to its anatomic structure. In addition, the lung is also exposed to various sources of endogenous OS produced via mitochondrial respiration and inflammation to viral and bacterial infections. The environmental sources of airborne OS involve oxidant gases and nanoparticles and ultrafine particles from car exhaust fumes and industrial pollution. Nonetheless, tobacco smoking is the most significant risk factor for COPD in the Western world. On the other hand, the inhalation of combustion products from enclosed cooking fires is a significant additional etiological factor in developing countries [79]. Although tobacco smoking can trigger the onset of COPD (Figure 1), once COPD has occurred, smoking cessation does not stop COPD progression and the continuous presence of OS [80]. Various endogenous sources including mitochondrial respiration mediate the continuous presence of OS. In the presence of carbonyl stress, airway epithelial cells trigger the generation of ROS derived from mitochondria. In COPD patients, airway smooth muscle cells generate an increased level of mitochondrial-derived ROS when exposed to inflammatory stress from interferon-gamma, tumor necrosis factor-alpha, and interleukin (IL)-1. It has been revealed by pathway studies that mitochondrial dysfunction around complexes I and III are closely linked with COPD (Figure 1) [81,82]. Furthermore, various other intracellular sources of ROS include several cytoplasmic ROS-generating enzymes including heme peroxidases, xanthine/XO system, and NOX. The levels of these enzymes were found to be increased in inflammatory cells within the airways of COPD patients [83]. Although profusely generated superoxide radicals are a comparatively weaker oxidizing agent, however, it is the precursor for various other more detrimental ROS species including hydroxyl radicals, which are increased in the case of COPD, or the highly damaging and strong ONOO radicals generated by the fast reaction of superoxide with nitric oxide [3,84,85]. In a similar manner, myeloperoxidase (MPO), secreted by activated neutrophils, is likely to build up in the lungs of COPD patients, which can further result in the generation of highly damaging HOCl. Nevertheless, intracellular antioxidant defenses are able to effectively clean up these ROS species in healthy cells, and can therefore limit their impact.

5. Role of Oxidative Stress in COPD Pathogenesis

A comparative deficit of antiproteases including alpha-1 antitrypsin (AAT) (because of their inactivation mediated via oxidants) may trigger the imbalance between protease and antiprotease in the lungs, which can further form the basis of the protease–antiprotease paradigm of emphysema pathogenesis [86]. Various studies have revealed the in vitro AAT inactivation via oxidants secreted from inflammatory leukocytes or oxidants from CS, but this activity is less common in vivo [87]. A protease–antiprotease imbalance including elastase and AAT is an overgeneralization, as other antiproteases and proteases are also likely to be involved. It has been reported that oxidant generating systems including xanthine/XO can induce the secretion of mucus from respiratory epithelium [88,89]. In addition, oxidants are associated with the signaling mechanisms for the epidermal growth factor receptor (EGFR), and EGFR plays a vital role in mucus secretion [90]. At low concentrations (100 μM), oxidants including HOCl or H2O2 can cause marked damage to ciliary beating and stasis [89,91]. Therefore, oxidant-induced hypersecretion of mucus and reduced mucociliary clearance might lead to mucus accumulation in the airways, which can eventually result in airflow limitation. Damage to the respiratory epithelium is a key early event that takes place following exposure to CS, which can be detected by an elevated level of the epithelial permeability of the airspaces. Both in vitro and in vivo studies have revealed that exposure to CS can lead to elevated epithelial permeability, which is partly reversible by antioxidants [92,93,94]. Intra and extracellular GSH seems to be essential in maintaining epithelial integrity following exposure to CS, as deficiency of lung GSH alone may result in an elevated level of in vitro and in vivo epithelial permeability of the airspaces [95,96]. Interindividual differences in antioxidant defenses might be a factor in defining whether COPD occurs in cigarette smokers.
It has been confirmed by biopsy studies that COPD is linked with an increased level of inflammation in the airways [97]. OS might be a process through which the inflammation of airspaces is increased in the case of COPD [98]. OS might also have a significant contribution in increasing inflammation via upregulating various redox-sensitive transcription factors including activator protein-1 (AP-1) and NF-κB. Furthermore, OS might also increase inflammation via upregulating various extracellular signal-regulated kinases including p38 mitogen-activated protein-kinase and c-Jun-N-terminal kinase signaling mechanisms. It has been reported that CS activates all of these aforesaid signaling mechanisms [99,100]. Genes for numerous inflammatory mediators are controlled via various oxidant-sensitive transcription factors including NF-κB [101,102,103]. Oxidants caused the secretion of multiple inflammatory mediators including NO, IL-1, and IL-8 in bronchial epithelial cells, alveolar epithelial cells, and macrophage cell lines, and these processes were found to be linked with an elevated level of expressions of the genes for these inflammatory mediators, and an elevated level of activation and the nuclear binding of NF-κB [104,105]. It has been observed that NF-κB can be activated and translocated to the nucleus in the lung tissues in COPD patients and cigarette smokers compared to healthy individuals [99,106,107].
Activation of NF-κB in lung tissues is associated with the forced expiratory volume in the first second (FEV1) [108]. Interestingly NF-κB linking to its consensus location in the nucleus can result in increased pro-inflammatory gene transcriptions, and as a result inflammation, which can further lead to the increased OS and can create a vicious circle of elevated levels of inflammation resulting from the elevated level of OS (Figure 2). The influx of neutrophils in the lungs was found to be linked with an elevated level of NF-κB activation and the expressions of IL-8 genes and protein secretion in animal models of smoke exposure [109]. Indeed, all of these aforementioned processes were reported to be linked with OS, as these processes can be abrogated via antioxidant therapy [110,111,112]. OS can also induce chromatin remodeling, which might increase the level of inflammation in the lungs, further permitting access for RNA polymerase and NF-κB to the transcriptional machinery to increase the gene expression, and this event is familiar as oxidant sensitive [98]. It is hypothesized in emphysema pathogenesis that the loss of lung cells (particularly endothelial cells) might take place as a primary mechanism in the emphysema development due to the apoptosis of endothelial cells [113]. An increased level of apoptosis has been observed in emphysematous lungs compared to the lungs of nonsmokers [114]. Vascular endothelial growth factor receptor 2 (VEGFR2) can also mediate endothelial apoptosis. VEGFR2 downregulation resulted in emphysema in animal models and such VEGFR2 downregulation has also been detected in emphysematous lungs [114]. Apoptosis/emphysema mediated by the suppression of VEGF in animal models was linked with elevated OS markers and was averted via antioxidants [113], further indicating that OS is associated with this event and systemic effects including muscle dysfunction in the case of COPD [115].

6. Pulmonary Strategies of Antioxidant Defense

6.1. Glutathione

Since the lungs are continuously exposed to endogenous and external sources of OS, they have created various effective antioxidant defensive approaches (Figure 1), where a decreased GSH level has a significant contribution. As a by-product of metabolism, as much as 20% of all generated GSH can be found within the mitochondria to counteract the production of endogenous ROS [116]. It has been revealed that the introduction of airway epithelial cells from healthy individuals to acute OS induces an elevated level of GSH synthesis via increasing the expression and function of glutamyl-cysteine ligase (an important enzyme in the synthesis of GSH) [117]. Nevertheless, the level of glutamyl-cysteine ligase decreases near the central bronchial epithelium and in alveolar macrophages in COPD patients and smokers, which indicates a faulty regulatory process [118]. Comparable differential reactions between the control individuals and COPD patients were obvious with various other GSH-dependent antioxidant enzymes including the glutathione-S-transferase (GST) pi isoenzyme, GSH peroxidase, and GSTM1 [119]. Moreover, a genetic deletion mutation in GSTM1 was liked with emphysema development in smokers and elevated vulnerability in COPD development [120]. In a similar manner, COPD has also been linked with the genetic polymorphisms in the GST pi isoenzymes [121].

6.2. Restoration or Upregulation of Nrf2 Function

In the airway smooth muscle cells, decreased levels in the expressions of various antioxidant enzymes including superoxide dismutase 2 (SOD2) and catalase were decreased, while the expression of transforming growth factor β (TGF-β) was elevated in the case of COPD [122]. These aforesaid antioxidant enzymes are important for counteracting mitochondrial-derived ROS and are controlled by the transcription factor forkhead box O-3 (FoxO3). It has been observed that there is a deficit in FOXO3 function in the case of COPD [123]. SOD2 gene polymorphism is also strongly linked with COPD; however, insufficient data are available to demonstrate how these polymorphisms are associated with the functions [124]. SOD3 polymorphisms have also been linked with decreased lung activity in the case of COPD and defense against COPD development in smokers when activities of SOD3 are increased [125,126]. Nuclear erythroid-2-related factor 2 (Nrf2) controls more than 200 cellular detoxification and antioxidant enzymes, which also control the expression of genes via binding with the antioxidant response elements within the promoters of numerous cytoprotective and antioxidant genes [127]. A decreased level of expressions of Nrf2-responsive genes because of decreased Nrf2 function has been reported in COPD patients [128]. Thus, the restoration or upregulation of Nrf2 function might be an effective therapeutic approach in the case of COPD [129].

7. Oxidative Stress Biomarkers in Chronic Obstructive Pulmonary Disease

7.1. Exhalation of NO

In the lungs, the gas NO is endogenously generated via NO synthase (NOS), which can exist in both the inducible isoform (iNOS) and constitutive isoform (cNOS). iNOS can be mediated via inflammatory stimuli in the lungs and might thus indicate airway inflammation. Thus, exhaled NO levels are regarded as an indirect measure of OS and a marker for airway inflammation [130,131]. Nonetheless, the findings on the usage of NO concentrations as a marker for COPD are questionable. In stable COPD patients, an increased level of NO was found in exhaled air whereas others had lower or normal levels of exhaled NO compared to the control individuals [132]. These inconsistencies in the findings might take place because of the different criteria for patient selection or the use of different approaches of measurement. In addition, NO is short-lived in vivo and by rapidly reacting with the superoxide, it can be transformed into NOx [133,134]. NO might also produce stable S-nitrothiols (RS-NOs) by interacting with low molecular weight thiols including N-acetylcysteine or GSH to increase its bioactivity [135,136,137]. Therefore, instead of NO, RS-NOs are considered as the major products of inflammation and NOS. Compared to the healthy control individuals and nonsmokers, reports on the RS-NOs concentrations in inflammatory airway diseases exhibited elevated concentrations in the exhaled breath condensate (EBC) of smokers and patients with COPD [130].

7.2. Exhalation of Hydrogen Peroxide

H2O2 in exhaled air signifies oxidant production in the lungs. It has been reported that COPD patients and smokers exhale H2O2 much higher than nonsmokers or ex-smokers with COPD [138,139]. The level of H2O2 in exhaled air is even higher during acute exacerbations of COPD. Although the exact cause of this increased exhalation of H2O2 is not fully known, this may partially take place from the elevated superoxide anion (O2) secretion by the alveolar macrophages from smokers than the alveolar macrophages from nonsmokers [140,141]. Compared to nonsmoking individuals, an increased level of intracellular iron has been observed in the pulmonary macrophages of smokers [142]. In the airspaces of smokers, the presence of elevated levels of free irons might elevate the production of even more ROS via the Fenton reaction [143]. The combination of xanthine/XO has the capacity to produce H2O2 and superoxide anion radicals. This combination was found to be elevated in the plasma and bronchoalveolar lavage (BAL) of smokers and COPD patients compared to nonsmokers and healthy individuals, respectively. Interestingly, patients with COPD performing strenuous exercise experienced systemic OS, which was suppressed via blocking XO [4].

7.3. Inflammatory Response

Multiple studies have assessed the contribution of ROS in the production of the inflammation that takes place in the peripheral and central airways of patients with COPD [100,144]. Common characteristics of lung inflammation include the activation and recruitment of neutrophils, and the activation of resident macrophages and epithelial cells [100]. Oxidants that are present in CS have the capacity of inducing alveolar macrophages to secrete various mediators, some of which engage neutrophils and various other inflammatory cells in the lungs [100,145,146,147,148]. In addition, elevated numbers of neutrophils and macrophages move into the lungs of smokers, where these cells produce ROS by the reduced NOX [149,150,151,152,153]. The lungs of smokers containing airway obstructions possess more neutrophils compared to smokers without airway obstruction [154,155]. During acute exacerbations, peripheral blood neutrophils from COPD individuals and smokers exhibit an elevated generation of superoxide anions. In patients with COPD, this generation level was found to be returned to normal when they were clinically stable [156,157]. It has been reported that the MPO level of the neutrophils is positively linked with tobacco smoking, indicating an elevated level of the generation of oxidants including hypochlorous acid in smoking individuals [83]. A connection between the FEV1 and circulating neutrophil numbers has also been observed, which further indicates an elevated level of airflow limitation due to the ROS generation of the elevated level of neutrophils [158]. An elevated level of ROS release takes place from the circulating neutrophils in tobacco smokers with COPD compared to tobacco smokers without COPD [159].

7.4. Lipid Peroxidation (LPO)

In biological tissues, ROS can induce the peroxidation of polyunsaturated fatty acids, which can lead to the transformation of fatty acids into lipid hydroperoxides. Then, lipid hydroperoxides and lipid peroxides can react with nonenzymatic or enzymatic antioxidants or decay after interacting with iron-containing proteins or metal ions, which can lead to the formation of unsaturated aldehydes and hydrocarbon gases as by-products [160,161]. Lipid peroxidation (LPO) measured as thiobarbituric acid-reactive substances showed an increase in concentrations in the lungs and breath condensate of stable COPD individuals [141,162]. Furthermore, these LPO products are negatively linked with the FEV1, which indicates that LPO has a significant contribution in deteriorating lung activity [163]. An increased level in the LPO product has been observed in the bronchoalveolar lavages and plasma of healthy smokers. Moreover, elevated LPO product levels are inversely linked to the extent of small airway obstruction and time expired from the last exposure to CS [164,165]. 4-Hydroxy-2,3-nonenal (an end-product of LPO) has the capacity to modify cellular proteins. The respiratory endothelial and epithelial cells of smokers with airway obstructions had increased levels of 4-hydroxy-2,3-nonenal-modified proteins in comparison with the individuals without airway obstruction or nonsmokers [166]. Ethane (a hydrocarbon) can be generated as a by-product of the peroxidation of various fatty acids including 9,12,15-linolenic acid [131]. Exhaled ethane is increased in the case of COPD patients compared to the control individuals. This elevated level is negatively associated with lung activity, which indicates that LPO is a crucial factor in COPD progression [131].

7.5. Protein Degradation

OS makes proteins more vulnerable to proteolytic degradation by altering amino acid chains, which results in the formation of protein aggregates and the cleavage of peptide bonds. Some of the amino acid residues are transformed to carbonyl residues during this process, which can be found systemically [167,168,169,170]. Following exposure to the gas phase of CS, human plasma proteins are altered to carbonyl-containing proteins with lost sulfhydryl groups. In CS, the unsaturated and saturated aldehydes play roles in modifying proteins [171,172]. It has been revealed by in vitro studies that the exposure of human plasma to CS resulted in raised levels of carbonyl proteins and depleted levels of plasma protein sulfhydryls [173]. OS-mediated damage of proteins and thus the generation of carbonyl proteins due to the exposure of CS were found to be partially prevented by GSH and completely by ascorbic acid [174]. Cigarette smoking can also lead to the formation of RNS, which can also cause the degradation of plasma proteins via oxidation and nitration [100]. Compared to nonsmokers, concentrations of oxidized proteins are considerably higher in smokers [175], and compared to nonsmokers, smoking individuals also showed increased levels of various nitrated proteins including plasminogen, ceruloplasmin, transferrin, and fibrinogen [175].
Aldehydes found in CS might interact with the amino and sulfhydryl moieties of plasma proteins through a Michael reaction [176,177]. The transformation of tyrosine into dityrosine and 3-nitrotyrosine can be considered as an indicator of protein and free radical damage [178,179]. The levels of nitrotyrosine were found to be increased in the epithelial lining fluid and plasma of tobacco smokers and negatively linked with the FEV1 [180]. Alpha 1-proteinase inhibitor (alpha 1-PI), an inhibitor of elastase activity, has a significant contribution in patients with COPD and its activity was reported to be reduced by oxidizing agents [181,182]. In alpha 1-PI, the oxidation of an important residue of methionine amino acid into methionine sulfoxide markedly reduced the suppressive ability of alpha 1-PI [170,183,184]. It has been revealed by BAL that smokers contain alpha 1-PI, which only exhibited half of its normal function, whereas alpha 1-PI from the lung washings of nonsmoking individuals is fully functional with only native methionine [141].

8. Strategies for Reducing Oxidative Stress by Antioxidants in the Treatment of COPD

8.1. Thiol-Based Antioxidants

8.1.1. Carbocysteine

S-carboxymethylcysteine (S-CMC or carbocysteine) (Figure 3) is a thiol derivative of L-cysteine. It has been revealed that S-CMC has anti-inflammatory, mucoactive, and antioxidant effects. Currently available oral preparations of carbocysteine include S-CMC and lysine salt of S-CMC (S-CMC-lys). In the gastrointestinal (GI) tract, the lysine residue in S-CMC-Lys breaks down to generate the active drug S-CMC. Compared to erdosteine and N-acetyl-L-cysteine (NAC), the mucoactive effect of carbocysteine is different from other thiol-based mucolytics as carbocysteine elevates the sialomucin level, which affects the rheological effects of mucus through the suppression of kinins [185]. Furthermore, carbocysteine mediates mucociliary clearance velocities, specifically in individuals with chronic bronchitis who have a lower level of clearance prior to treatment [185]. Carbocysteine also provided protection against emphysema triggered by CS in the rat models [186]. In patients with COPD, the treatment with S-CMC-Lys for a duration of 6 months markedly reduced the concentrations of the 8-isoprostane (an LPO product) and pro-inflammatory cytokine including IL-6, which suggests that S-CMC-Lys showed both antioxidant and anti-inflammatory effects [187]. Since this drug can also decrease the levels of bacterial respiratory tract infections in the case of COPD [47], carbocysteine might therefore play a role by suppressing the adherence of pathogens to the cells.
Compared to the placebo-treated group, the in vitro studies revealed that treatment with carbocysteine can decrease the attachment of Moraxella catarrhalis (a bacterium to pharyngeal epithelial cells in individuals with chronic bronchitis and healthy subjects [188,189,190]. Carbocysteine can also markedly decrease the adherence of Streptococcus pneumoniae to pharyngeal epithelial cells [47]. In COPD patients, carbocysteine can decrease the occurrence of common colds and related exacerbations, a characteristic that can contribute to its capacity to reduce the expression of intercellular adhesion molecule 1 in the respiratory tract [191]. The effect of treatment with carbocysteine (250 mg, 3 times a day) has been assessed for 3 years in 709 Chinese COPD patients. It was observed in a clinical study that COPD patients receiving carbocysteine faced less frequent exacerbations per year [192]. Moreover, most of these COPD patients were not administered corticosteroids [192]. The clinical trials of carbocysteine in COPD patients are summarized in Table 3.

8.1.2. N-Acetyl-L-Cysteine

NAC (Figure 3) is a powerful reducing agent and an acetyl derivative of the amino acid. NAC has mucolytic properties and it decreases the viscosity of mucus, thus ameliorating mucociliary clearance. On the other hand, in the GI tract, NAC is deacetylated to cysteine, which plays a role as a GSH precursor. NAC has the capacity to neutralize oxidant species by reducing disulfide bonds. In lungs, NAC can also elevate the level of intracellular GSH in vivo as it can reduce intracellular cystine to cysteine. Among the thiol derivatives, NAC has been extensively studied both in vivo and in vitro. Oral administration of NAC decreased the level of elastase-induced emphysema in rat models [198]. Furthermore, NAC provided protection against the tobacco smoke-mediated oxidation of the Z variant of AAT in early-onset emphysema in mouse models [199]. Since GSH is an important lung antioxidant, NAC has primarily been utilized to increase the level of lung GSH in COPD patients [200,201]. In Table 4, the clinical trials on the benefits of using NAC in COPD patients have been summarized.
In a clinical study, NAC treatment at the dose of 600 mg twice per day for 6 months decreased several BAL fluid and plasma oxidative biomarkers in tobacco smokers [210]. On the other hand, NAC treatment at the dose of 600 mg two times per day for 2 months decreased the oxidative burden in the airways of individuals with stable COPD [207], and was linked with ameliorated lung symptoms and decreased risk of exacerbation in people with chronic bronchitis [206]. In severe COPD patients, NAC ameliorated the muscle activity by increasing the time of quadriceps endurance, along with a reduction in the systemic OS markers [211]. Various reports have also shown reduced levels of exacerbations by 29% [212,213]. In a large multicenter trial, NAC did not affect the decline in FEV1 or the frequency of the exacerbations [203]. Moreover, in this study, a reduced level of exacerbation frequency and overinflation was observed in COPD patients who did not receive inhaled glucocorticoids [203]. In the GI tract, NAC needs to be deacetylated to cysteine to play a role as a GSH precursor and is not highly bioavailable to elevate the level of GSH. Therefore, in order to identify any clinical advantage in COPD treatment, more studies are needed to evaluate the effects of NAC at higher doses (1200 mg or 1800 mg daily) or evaluate various other thiol derivatives that show higher bioavailability.

8.1.3. Fudosteine

Fudosteine (Figure 3) is used as an antioxidant and mucolytic agent. Compared to NAC, fudosteine has higher bioavailability and plays a role as an antioxidant by elevating the level of intracellular cysteine. Fudosteine also suppressed the hypersecretion of mucin by downregulating the gene expression of Mucin 5AC (MUC5AC) [214]. Fudosteine decreased the expressions of p-ERK in in vitro bronchial epithelial cell lines and in vivo expressions of p-ERK and phospho-p38 MAPK [214]. In lung epithelial cells, fudosteine suppressed the airway nitrative stress induced by ONOO by directly scavenging ONOO [215]. Therefore, as a mucoactive agent, fudosteine might be useful in the therapy of various chronic respiratory diseases such as bronchiectasis, COPD, chronic bronchitis, and bronchial asthma [214,216].

8.1.4. Erdosteine

Erdosteine (Figure 3) is a thiol-based antioxidant and mucoactive agent. Erdosteine was mainly utilized as a mucolytic agent. It breaks the disulfide bonds of mucus glycoproteins and affects the physical properties of the mucus, which can further result in an elevated level of mucus clearance [217]. In addition, erdosteine has antibacterial, anti-inflammatory, and antioxidant properties. In a clinical trial, the oral administration of erdosteine (300 mg twice daily) for 8 months markedly reduced exacerbations and improved the quality of health in comparison with the placebo [218]. Erdosteine can also be beneficial for people suffering from severe, prolonged, or repeated COPD exacerbations [219,220]. Erdosteine also decreased the frequency of severe exacerbations, necessitating hospital admissions [217]. Erdosteine (300 mg two times a day) administration for 7–10 days ameliorated symptoms and decreased the length of hospitalization in individuals with exacerbations of COPD [221]. The administration of erdosteine (600 mg daily) with procysteine ameliorated the concentrations of various chemotactic cytokines including IL-6 and IL-8 and improved the concentration of CS-mediated ROS generation via alveolar macrophages in the bronchial secretions of tobacco smokers with COPD [220]. In COPD patients, erdosteine also reduced the concentrations of pro-inflammatory eicosanoids in the blood of patients with COPD [222].

8.2. Superoxide Dismutase Mimetics

There are three classes of SOD mimetics. The first class of SOD mimetics involves various manganese (Mn)-based macrocyclic ligands including M40419, M40403, and M40401 [113,223]. The second category involves multiple Mn-metaloporphyrins including AEOL-10150 and AEOL-10113 226,262], whereas the third class includes salens (Mn-based SOD mimetics). Since salens also have a catalase-like function, they can also neutralize ONOO and H2O2 [224]. In animal models of airway inflammation, so far, only the second category of SOD mimetics has been investigated. In rat models, AEOL10150 (a SOD mimetic) suppressed the tobacco smoke-mediated lung inflammation, LPO, and the production of ONOO [225]. It has been observed that treatment with recombinant SOD can reduce the cigarette smoke-induced release of IL-8 and prevent the influx of neutrophils into the lungs, which further indicates its potential as an anti-inflammatory and antioxidant in the case of COPD [95]. MnTE-2-PyP (a Mn porphyrin) is an antioxidant and ROS scavenger. MnTE-2-PyP has been reported to have the capacity to scavenge H2O2, ONOO, lipid peroxides, and superoxide [226].
Treatment with MnTE-2-PyP decreased damage and inflammation-mediated via numerous factors [227,228]. The MnTE-2-PyP-mediated anti-inflammatory properties mainly take place because of its capacity to decrease MnTE-2-PyP. Collectively, these findings suggest that these compounds might have the potential to treat COPD. In the lungs, extracellular SOD (ECSOD or SOD3) is found to be highly expressed. ECSOD is found in the blood vessel linings of the lungs, the surface of the airway smooth muscle, and the extracellular matrix in the junctions of the airway epithelial cells [229]. SOD3 also has the capacity to directly scavenge O2•−, thus it has a significant contribution in providing protection against OS in the lung. Furthermore, SOD3 can reduce CS-mediated OS in mouse macrophages [230]. Moreover, SOD3 decreases emphysema and lung inflammation by reducing the oxidative fragmentation of the extracellular matrix (ECM) including elastin and heparin sulfate [231]. Thus, the discovery of therapeutic agents to increase or replenish the level of SOD3 in the lungs might be an effective therapeutic intervention for emphysema/COPD.

8.3. Nrf2 Activators

Nuclear factor erythroid-2-related factor 2 (Nrf2) is found in the cytoplasm of normal cells. This transcription factor has a significant contribution in providing protection against ROS and electrophiles. As a reaction to electrophilic stress and OS, Nrf2 detaches from Kelch-like ECH-associated protein 1 (Keap1) to translocate into the nucleus, where it has been reported to bind with the antioxidant response element (ARE) of target genes [232,233,234]. It has been observed that Nrf2 controls nearly all of the phase II cytoprotective genes and antioxidants, for instance, glutathione peroxidase (GPx), glutamate-cysteine synthase, glutamate-cysteine ligase modifier subunit (GCLM), NAD(P)H/quinone oxidoreductase 1 (NQO1), and multiple glutathione S-transferase family members [232]. Nrf2 knockout mouse models exhibited an elevated level of susceptibility to CS-mediated emphysema compared to wild-type mouse models, which suggests a protective function of Nrf2 [235,236].
DJ-1 (a Nrf2 stabilizer) loss and post-translational modification of the Keap1–Bach1 equilibrium can lead to Nrf2 downregulation in the lungs of COPD patients [237,238,239,240]. CDDO-imidazolide (a Nrf2 activator) was found to provide protection against CS-mediated emphysema in mouse models [234]. Nrf2 activation via sulforaphane (found in cruciferous vegetables and broccoli) can result in reducing some of the biochemical changes that take place in COPD patients and smokers [241]. Chalcones show anti-inflammatory properties because of their capacity to suppress the NF-κB signaling pathway [242,243] and activate the Nrf2/ARE signaling pathway therefore triggering expressions of phase II detoxifying enzymes [244]. Multiple chalcone derivatives are also being invented that are likely to have the potential to treat COPD [245]. Nevertheless, the toxicity, bioavailability, and pharmacokinetics of these compounds in the lungs are yet to be fully revealed.

8.4. NOX Inhibitors

In COPD, NOX (a membrane-bound complex) plays a role as an important source of ROS through the production of superoxide anions. There are various isoforms of the catalytic component of NOX such as dual oxidases (Duox1 and Duox2) and NOX1-5 [246]. Various inhibitors of NOX have also been invented to counter OS [247,248]. In CS-exposed mouse models, systemic administration of apocynin (a non-selective inhibitor of NOX) decreased the inflammatory chemokines and cytokines in BAL fluid [249]. Nebulized apocynin administration decreased the levels of nitrite and H2O2 in the exhaled air in patients with COPD, however, no clinical parameters were observed [250]. In addition to other functions, various polyphenols including resveratrol and quercetin suppressed the NOX function. Indeed, it is difficult to develop highly selective inhibitors of NOX. Setanaxib is a dual inhibitor of Nox1/4 that decreases acute lung damage mediated via reperfusion injury, however, this effect is yet to be confirmed in COPD models and clinical trials [251].

8.5. Antioxidant Mimetics

Antioxidant mimetics have been created to reinstate the depleted levels of endogenous antioxidants including GPx, catalase, and SOD [252]. SOD mimetics involve various metalloporphyrins including AEOL 10150, AEOL 10113, and Mn-containing molecules including M40419. These SOD mimetics were found to be effective in several animal models of OS, for example, CS-exposed mouse models showed a decreased level of inflammation [225]. AEOL 10150 is mainly used to treat radiation pneumonitis and the development of its derivatives is ongoing for the treatment of patients with COPD. GPx includes non-selenium and selenium-containing antioxidant enzymes that catalyze H2O2 breakdown. The level of GPx-1 is decreased in the plasma and lungs of COPD patients, which indicates that GPx-1 mimetics might be therapeutically beneficial [253]. GPx transgenic mouse models have protection against emphysema and inflammation following CS exposure, while GPx gene knockout elevated the lung response to CS [254]. Ebselen is a GPx mimetic and a selenium-based organic complex. This GPx mimetic is a strong antioxidant and it significantly neutralizes peroxynitrite radicals [255]. Ebselen suppressed the NF-κB/AP-1 activation, and therefore the expressions of pro-inflammatory genes in human leukocytes treated with peroxynitrite. Ebselen effectively decreased the ozone-induced airway inflammation in rat models [256] and inflammatory cytokines in the lungs of CS-exposed mouse models [249], however, no clinical trials have been carried out in COPD patients.

8.6. iNOS Inhibitors and Spin Trapping Agents

It has been demonstrated that iNOS suppression via several chemical inhibitors including NG-nitro-L-arginine methyl ester and L-N6-(1-Iminoethyl)lysine (L-NIL) weakened animal models of pulmonary emphysema [257,258]. In addition to supplementation with other antioxidants, selective iNOS inhibition might be an effective approach in managing COPD [259]. Spin trapping agents are chemicals that can cause the quenching of free radicals to generate measurable stable end-products. Most of the spin trapping agents contain a nitroxide- or nitrone-nucleus and are derivatives of these moieties. Spin trapping agents have been extensively investigated in vitro. Their therapeutic potential has also been investigated in in vivo animal models of lung inflammation by utilizing α-phenyl-N-tert-butyl nitrone [260,261,262]. Spin trapping agents that were created earlier had very short half-lives and produced toxic hydroxyl radicals on decay. However, this problem has been solved by introducing electron-withdrawing moieties around the core of the pyrroline rings [263]. Azulenyl- and isoindole-based nitrones including azulenyl nitrone (STANZ) show potent antioxidant effects and have the capacity to suppress in vitro LPO [264,265]. On the other hand, derivatives of phenyl-base nitrone spin trap including NXY-059 exerted beneficial effects in several animal models of lung diseases [266,267,268].

8.7. Suppression of Nitrative Stress

Superoxide anions rapidly combine with NO to generate extremely reactive peroxynitrite ions, which can further lead to the generation of 3-nitrotyrosine adducts of amino acids in proteins that might disturb their activities as structural proteins, ion channels, or enzymes. The level of peroxynitrite is elevated in the EBC of patients with COPD [215], while 3-nitrotyrosine is expressed in the airways and sputum cells of COPD individuals [269,270]. The generation of peroxynitrite in the airways of patients with COPD might elucidate why the level of fractional exhaled NO (FeNO) is low in COPD individuals, since the entire free NO is closely bound by superoxide anions. NO is expressed in the alveolar epithelial cells. In response to OS, NO might be produced in the lungs of COPD patients by NOS1 [258]. Mouse models exposed to CS showed an elevated level in the expression of iNOS (NOS2) and were found to be protected against emphysema development by using selective inhibitors of iNOS and via knockout of the iNOS gene [271]. When aminoguanidine (a comparatively selective iNOS inhibitor) was administered through nebulization, it partly decreased peripheral and central exhaled levels of NO in COPD individuals, however, it failed to remove exhaled NO, indicating that NOS1 is the probable cause and those selective inhibitors of iNOS might not be beneficial in decreasing the level of peroxynitrite in patients with COPD [258]. Various highly selective inhibitors of iNOS have been invented for clinical administration. L-NIL (a selective inhibitor of iNOS) is highly effective in decreasing the level of FeNO in asthma patients; however, it is yet to be studied in patients with COPD [272].

8.8. Lazaroids and Edaravone

Lazaroids are a group of non-glucocorticoid analogs of methylprednisolone and they have the capacity to cross hydrophobic areas of the cell membrane, particularly to avert membrane LPO [261,273,274]. Their protective actions have already been demonstrated in numerous animal models of lung injury [274,275]. Lazaroids mainly exert their protective actions by suppressing LPO. Lazaroids suppressed the secretion of TNF-α from alveolar macrophages and the generation of free radicals [276,277]. However, more studies are needed to assess the effectiveness of lazaroids as a therapeutic agent in COPD treatment. Edaravone is an LPO inhibitor and a strong scavenger of protein carbonyls and free radicals [278,279]. Since carbonyl stress and protein carbonylation via aldehydes are present in COPD, edaravone can provide protection to the lungs against the activities of these oxidative products [280,281]. Moreover, edaravone improved the OS, inflammation, lung damage, and even mortality, mediated by intestinal ischemia-reperfusion in the rat models [282]. Collectively, all of these findings suggest that edaravone can be effective in COPD treatment.

8.9. Enzymatic and Small Molecule Antioxidants

Various antioxidant enzymes including GPx, catalase, and SOD can effectively neutralize cellular ROS. However, the functions and expressions of these antioxidant enzymes are changed in various diseases including OS. Reinstatement of altered antioxidant enzyme function can be attained via small molecules having catalytic effects, which can mimic the function of the enzyme. Various small molecule antioxidants have been created, however, only a few of them have been investigated in clinical studies. Various nitrone spin trap antioxidants including disufentan sodium were created as a neuroprotective agent, however it failed in clinical studies of acute stroke. Thioredoxin (Trx) (an endogenous regulator of redox balance) has the capacity to neutralize and its level might be decreased in COPD [283]. Systemic Trx-1 administration was found to be effective in reducing the neutrophilic inflammation in murine models of COPD [284]. Redox effector factor-1 (Ref-1) and Trx belong to the oxidoreductase family of redox sensors. Trx has the capacity to bind to various proteins including apoptosis signal-regulating kinase and hepatopoietin, which are secreted from these complexes during OS [285].
Following dissociation, Trx was found to reduce an important key thiol group within the p65/NF-κB subunit, which can further result in transcriptional activation [286,287,288]. In an animal model, suppression of Trx by MOL-294 (a small molecule Trx inhibitor) blocked the nuclear activation of AP-1-, and NF-κB-dependent transcription led to a reduced influx of neutrophils and the generation of TNF-α [289]. Trx activation through small molecules decreased the level of OS [290]. Trx-1 overexpression, mainly because of its antioxidant effect, decreased CS-induced OS and emphysema [291], but the actions in COPD have yet to be evaluated. It has been observed that XO might produce superoxide anions and is elevated in the lungs of mouse models exposed to CS [292]. An increased level in the expression of XO was observed in the bronchial mucosal lining fluid of patients with COPD and is linked with the elevated expressions of various proinflammatory cytokines [293]. Allopurinol (a XO inhibitor) (Figure 3) decreased the expression of 3-nitrotyrosine in the sputum cells of patients with COPD and elevated the level of FeNO, perhaps by blocking the superoxide production so that ONOO− is not generated [294].

8.10. Dietary Antioxidants

Diets that are poor in antioxidants have been linked to poor lung activity and can be a risk factor for COPD development, however, it is challenging to demonstrate that antioxidant vitamins precisely ameliorate established COPD [295]. There are various dietary antioxidants including flavonoids (e.g., quercetin), resveratrol (a dietary polyphenol), vitamin E, and vitamin C, however, improving the intake of dietary antioxidants has not been confirmed to ameliorate clinical features of COPD or lung activity [296,297]. It has been reported that the Mediterranean diet contains an increased level of dietary antioxidants and some evidence suggests that it might provide protection against COPD development; however, factors including poverty are challenging to separate out [298]. Resveratrol present in red wine and red-skinned fruits show in vitro anti-inflammatory and antioxidant properties in COPD cells and decreases lipopolysaccharide-induced pulmonary neutrophilic inflammation in rat models [299,300]. Nonetheless, its oral bioavailability is poor, therefore, stronger and more orally bioavailable analogs have been developed. Resveratrol, through inhaled formulations, decreased the enhanced lung aging in telomerase-deficient mice [301]. It has been reported that (-)-epigallocatechin (a polyphenol present in green tea) can cause the activation of FOXO3a, which is a transcription factor that controls various antioxidant genes including catalase and SOD [302].

8.11. Peroxidase Inhibitors

An increased level of neutrophil-derived MPO has been observed in the case of COPD, which reflects the activation of neutrophils in the lungs [303]. So far, multiple inhibitors of MPO have been created [304]. AZD5904 (a selective and irreversible inhibitor of MPO) decreases OS and emphysema development in CS-exposed guinea pigs [305]. Even though this drug was well-tolerated in human participants, it has been discontinued for unknown causes.

8.12. Mitochondria-Targeted Antioxidants

It has been confirmed that dysfunctional mitochondria are present in the case of COPD [306]. Mitochondrial mass was found to increase, along with the disruption and fusion of mitochondria with leakiness of the mitochondrial membranes [307]. This can be partially clarified by weakened autophagy processes that eradicate damaged mitochondria (mitophagy). In the case of COPD, dysfunctional leaky mitochondria might serve as the main source of ROS [308,309]. There are various available drugs that can particularly target mitochondria [310,311,312]. Mitochondria-targeted antioxidants have been developed primarily based on ubiquinone’s structure, which can be 50- to 100-fold concentrated in the mitochondrial matrix. Furthermore, they were found to be more potent than multiple animal models of aging [311,313]. A number of mitochondria-targeted antioxidants including SkQ1, pyrroloquinoline quinone, mito-TEMPO, and mitoQ are currently being studied in several clinical studies for age-linked diseases. mito-TEMPO also suppressed the mROS secretion and mitochondrial dysfunction induced by CS in the human airway epithelial cells in vitro [314]. Treatment with mitoQ also decreased lung inflammatory mediators, neutrophilic inflammation, and airway hyperresponsiveness in a murine model of the chronic OS that involved long-term exposure to ozone [308].

9. Strategies to Improve Pulmonary Bioavailability of Antioxidants

Indeed, the beneficial actions of antioxidants largely depend on their pharmacokinetic properties. Therefore, the bioavailability of antioxidants needs to be studied before assessing their antioxidant properties [315]. Poor pulmonary bioavailability has limited the use of antioxidants in COPD treatment [139,316]. There are various factors including host metabolism, release pattern from foods, and absorption, which can affect the bioavailability of exogenous and dietary antioxidants [315]. Host and dietary factors can markedly limit the half-life, concentration, and absorption of exogenous antioxidants. Therefore, it is important to address these factors to develop strong antioxidants that would effectively maintain constant concentrations within lung tissue compartments [315,317].
Modifying existing antioxidants or developing new antioxidants that are resistant to phase II modification would be able to retain their bioavailability and potency, even after modification via enzymes. Tempol, a novel membrane-permeable radical scavenger, is effective in decreasing lung inflammation in response to shock [318,319]. Ameliorated inhaled delivery approaches have been developed to avoid the first pass metabolism that takes place during systemic absorption and to mediate sufficient levels of antioxidants to be reached in the lung [320,321]. Currently available inhalational devices are able to deliver most of the drugs only to medium and large size airways of the lung, while ignoring the alveolar regions and small airways that are major areas in the disease pathogenesis [322]. In addition, ameliorations in distal lung deposition are necessary before utilizing this technology for the effective delivery of antioxidants. In the near future, this might be achievable, since there are effective inhalational techniques that can mediate uniform distribution in the distal lung [323,324]. Future improvements in antioxidant therapy might include counteracting the CS-mediated stimulation of various oxidant-forming enzymes including xanthine oxidase and NOX, which are found within the lung epithelium. Moreover, these enzymes can play significant roles in OS, which is mediated by exposure to CS [325,326,327,328].

10. Future Directions

There is growing evidence that suggests that elevated ROS generation takes place in the case of COPD, which might be crucial for COPD pathogenesis. Various small molecules are currently being investigated in clinical studies that quench oxidants or target oxidant signaling induced by CS. In order to address these issues of COPD, various anti-inflammatory agents and/or antioxidants including antioxidant mimetics, dietary polyphenols, spin trapping agents, thiol molecules, and inhibitors of OS-mediated signaling pathways have been developed. In addition, antioxidants might increase the effectiveness of glucocorticoids by quenching aldehydes and oxidants, which further increases the histone deacetylase function in patients with COPD. Various dietary polyphenols including curcumin and resveratrol can also suppress the release of proinflammatory cytokines, histone acetylation, oxidant/CS-induced activation of NF-κB, and restore the functions of glucocorticoids by upregulating the function of histone deacetylase. Therefore, dietary polyphenols control inflammation at the molecular level, which might further restore the functions of glucocorticoids in the treatment of CS-mediated chronic inflammatory diseases. Thus, a potent and effective antioxidant with good bioavailability is important to regulate inflammatory and localized oxidative mechanisms that are present in COPD pathogenesis [329].

11. Conclusions

Indeed, OS and carbonyl stress play important roles in COPD pathogenesis. Targeting OS with potent antioxidants or increasing the endogenous antioxidants may be beneficial in the treatment of COPD. Antioxidants might also affect various issues of COPD including ECM remodeling, inflammation, the hypersecretion of mucus, and overcoming steroid resistance. Various small molecule antioxidants have been evaluated in preclinical and clinical studies. Even though thiol-based antioxidants can effectively target the ROS and cellular responses induced by oxidants, novel and more potent antioxidants with good bioavailability that can be used in COPD treatment need to be studied in clinical studies. Nevertheless, a limited number of clinical trials have been conducted and there is a deficiency of data regarding the toxicity, bioavailability, pharmacokinetics, and absorption of multiple activators of endogenous antioxidants and exogenous antioxidants.

Funding

This research was funded by Deputyship for Research and Innovation, Ministry of Education in Saudi Arabia by grant number RUP-5.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The author extends their appreciation to the Deputyship for Research and Innovation, Ministry of Education in Saudi Arabia for funding this research work through project number RUP-5.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lindberg, A.; Lindberg, L.; Sawalha, S.; Nilsson, U.; Stridsman, C.; Lundbäck, B.; Backman, H. Large underreporting of COPD as cause of death-results from a population-based cohort study. Respir. Med. 2021, 186, 106518. [Google Scholar] [CrossRef] [PubMed]
  2. Rahman, I. The role of oxidative stress in the pathogenesis of COPD: Implications for therapy. Treat. Respir. Med. 2005, 4, 175–200. [Google Scholar] [CrossRef] [PubMed]
  3. Thimmulappa, R.K.; Chattopadhyay, I.; Rajasekaran, S. Oxidative stress mechanisms in the pathogenesis of environmental lung diseases. Oxidative Stress Lung Dis. 2019, 2, 103–137. [Google Scholar] [CrossRef]
  4. Rahman, I.; Adcock, I.M. Oxidative stress and redox regulation of lung inflammation in COPD. Eur. Respir. J. 2006, 28, 219–242. [Google Scholar] [CrossRef]
  5. Yoshida, T.; Tuder, R.M. Pathobiology of cigarette smoke-induced chronic obstructive pulmonary disease. Physiol. Rev. 2007, 87, 1047–1082. [Google Scholar] [CrossRef] [PubMed]
  6. Anderson, D.; Macnee, W. Targeted treatment in COPD: A multi-system approach for a multi-system disease. Int. J. Chron. Obstruct. Pulmon. Dis. 2009, 4, 321–335. [Google Scholar] [CrossRef] [PubMed]
  7. Nishida, M.; Maruyama, Y.; Tanaka, R.; Kontani, K.; Nagao, T.; Kurose, H. Gα(i) and Gα(o) are target proteins of reactive oxygen species. Nature 2000, 408, 492–495. [Google Scholar] [CrossRef] [PubMed]
  8. Poli, G.; Leonarduzzi, G.; Biasi, F.; Chiarpotto, E. Oxidative Stress and Cell Signalling. Curr. Med. Chem. 2004, 11, 1163–1182. [Google Scholar] [CrossRef]
  9. Aoshiba, K.; Nagai, A. Senescence hypothesis for the pathogenetic mechanism of chronic obstructive pulmonary disease. In Proceedings of the American Thoracic Society; 2009; Volume 6, pp. 596–601. [Google Scholar]
  10. Tsuji, T.; Aoshiba, K.; Nagai, A. Alveolar cell senescence in patients with pulmonary emphysema. Am. J. Respir. Crit. Care Med. 2006, 174, 886–893. [Google Scholar] [CrossRef]
  11. Churg, A.; Wright, J.L. Proteases and emphysema. Curr. Opin. Pulm. Med. 2005, 11, 153–159. [Google Scholar] [CrossRef]
  12. Cavarra, E.; Lucattelli, M.; Gambelli, F.; Bartalesi, B.; Fineschi, S.; Szarka, A.; Giannerini, F.; Martorana, P.A.; Lungarella, G. 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. [Google Scholar] [CrossRef] [PubMed]
  13. Rahman, I.; Swarska, E.; MacNee, W.; Stolk, J.; Henry, M. Is there any relationship between plasma antioxidant capacity and lung function in smokers and in patients with chronic obstructive pulmonary disease? Thorax 2000, 55, 189–193. [Google Scholar] [CrossRef]
  14. Ahmad, A.; Shameem, M.; Husain, Q. Relation of oxidant-antioxidant imbalance with disease progression in patients with asthma. Ann. Thorac. Med. 2012, 7, 226. [Google Scholar] [CrossRef] [PubMed]
  15. Singh, S.; Verma, S.K.; Kumar, S.; Ahmad, M.K.; Nischal, A.; Singh, S.K.; Dixit, R.K. Evaluation of Oxidative Stress and Antioxidant Status in Chronic Obstructive Pulmonary Disease. Scand. J. Immunol. 2017, 85, 130–137. [Google Scholar] [CrossRef] [PubMed]
  16. MacNee, W. Pathogenesis of Chronic Obstructive Pulmonary Disease. Proc. Am. Thorac. Soc. 2005, 2, 258. [Google Scholar] [CrossRef] [PubMed]
  17. Rajendrasozhan, S.; Yao, H.; Rahman, I. Current perspectives on role of chromatin modifications and deacetylases in lung inflammation of COPD. COPD 2009, 6, 291. [Google Scholar] [CrossRef]
  18. Macdonald, J.; Galley, H.F.; Webster, N.R. Oxidative stress and gene expression in sepsis. Br. J. Anaesth. 2003, 90, 221–232. [Google Scholar] [CrossRef]
  19. Laniado-Laborin, R. Smoking and Chronic Obstructive Pulmonary Disease (COPD). Parallel Epidemics of the 21st Century. Int. J. Environ. Res. Public Health 2009, 6, 209. [Google Scholar] [CrossRef]
  20. Bhatt, S.P.; Kim, Y.I.; Harrington, K.F.; Hokanson, J.E.; Lutz, S.M.; Cho, M.H.; DeMeo, D.L.; Wells, J.M.; Make, B.J.; Rennard, S.I.; et al. Smoking duration alone provides stronger risk estimates of chronic obstructive pulmonary disease than pack-years. Thorax 2018, 73, 414–421. [Google Scholar] [CrossRef]
  21. Liu, S.; Zhou, Y.; Wang, X.; Wang, D.; Lu, J.; Zheng, J.; Zhong, N.; Ran, P. Biomass fuels are the probable risk factor for chronic obstructive pulmonary disease in rural South China. Thorax 2007, 62, 889–897. [Google Scholar] [CrossRef] [Green Version]
  22. Capistrano, S.J.; van Reyk, D.; Chen, H.; Oliver, B.G. Evidence of Biomass Smoke Exposure as a Causative Factor for the Development of COPD. Toxics 2017, 5, 36. [Google Scholar] [CrossRef] [PubMed]
  23. Sahni, S.; Talwar, A.; Khanijo, S.; Talwar, A. Socioeconomic status and its relationship to chronic respiratory disease. Adv. Respir. Med. 2017, 85, 97–108. [Google Scholar] [CrossRef] [PubMed]
  24. Grigsby, M.; Siddharthan, T.; Chowdhury, M.A.H.; Siddiquee, A.; Rubinstein, A.; Sobrino, E.; Miranda, J.J.; Bernabe-Ortiz, A.; Alam, D.; Checkley, W. Socioeconomic status and COPD among low- and middle-income countries. Int. J. Chron. Obstruct. Pulmon. Dis. 2016, 11, 2497. [Google Scholar] [CrossRef]
  25. Kanervisto, M.; Vasankari, T.; Laitinen, T.; Heliövaara, M.; Jousilahti, P.; Saarelainen, S. Low socioeconomic status is associated with chronic obstructive airway diseases. Respir. Med. 2011, 105, 1140–1146. [Google Scholar] [CrossRef] [PubMed]
  26. de Matteis, S.; Jarvis, D.; Darnton, A.; Hutchings, S.; Sadhra, S.; Fishwick, D.; Rushton, L.; Cullinan, P. The occupations at increased risk of COPD: Analysis of lifetime job-histories in the population-based UK Biobank Cohort. Eur. Respir. J. 2019, 54, 1900186. [Google Scholar] [CrossRef]
  27. Blanc, P.D.; Iribarren, C.; Trupin, L.; Earnest, G.; Katz, P.P.; Balmes, J.; Sidney, S.; Eisner, M.D. Occupational exposures and the risk of COPD: Dusty trades revisited. Thorax 2009, 64, 6–12. [Google Scholar] [CrossRef] [PubMed]
  28. Senn, O.; Russi, E.W.; Imboden, M.; Probst-Hensch, N.M. α1-Antitrypsin deficiency and lung disease: Risk modification by occupational and environmental inhalants. Eur. Respir. J. 2005, 26, 909–917. [Google Scholar] [CrossRef]
  29. DeMeo, D.L.; Silverman, E.K. α1-Antitrypsin deficiency · 2: Genetic aspects of α1-antitrypsin deficiency: Phenotypes and genetic modifiers of emphysema risk. Thorax 2004, 59, 259–264. [Google Scholar] [CrossRef]
  30. Zhang, Y.; Wang, L.; Mutlu, G.M.; Cai, H. More to Explore: Further Definition of Risk Factors for COPD—Differential Gender Difference, Modest Elevation in PM2.5, and e-Cigarette Use. Front. Physiol. 2021, 12, 423. [Google Scholar] [CrossRef]
  31. Ntritsos, G.; Franek, J.; Belbasis, L.; Christou, M.A.; Markozannes, G.; Altman, P.; Fogel, R.; Sayre, T.; Ntzani, E.E.; Evangelou, E. Gender-specific estimates of COPD prevalence: A systematic review and meta-analysis. Int. J. Chron. Obstruct. Pulmon. Dis. 2018, 13, 1507. [Google Scholar] [CrossRef] [Green Version]
  32. Shukla, S.D.; Shastri, M.D.; Jha, N.K.; Gupta, G.; Chellappan, D.K.; Bagade, T.; Dua, K. Female gender as a risk factor for developing COPD. EXCLI J. 2021, 20, 1290. [Google Scholar] [CrossRef] [PubMed]
  33. Sørheim, I.C.; Johannessen, A.; Gulsvik, A.; Bakke, P.S.; Silverman, E.K.; DeMeo, D.L. Gender differences in COPD: Are women more susceptible to smoking effects than men? Thorax 2010, 65, 480–485. [Google Scholar] [CrossRef] [PubMed]
  34. Tian, P.; Wen, F. Clinical significance of airway mucus hypersecretion in chronic obstructive pulmonary disease. J. Transl. Intern. Med. 2015, 3, 89. [Google Scholar] [CrossRef]
  35. Miravitlles, M. Cough and sputum production as risk factors for poor outcomes in patients with COPD. Respir. Med. 2011, 105, 1118–1128. [Google Scholar] [CrossRef]
  36. Zanini, A.; Cherubino, F.; Zampogna, E.; Croce, S.; Pignatti, P.; Spanevello, A. Bronchial hyperresponsiveness, airway inflammation, and reversibility in patients with chronic obstructive pulmonary disease. Int. J. Chron. Obstruct. Pulmon. Dis. 2015, 10, 1155–1161. [Google Scholar] [CrossRef]
  37. De Marco, R.; Accordini, S.; Marcon, A.; Cerveri, I.; Antó, J.M.; Gislason, T.; Heinrich, J.; Janson, C.; Jarvis, D.; Kuenzli, N.; et al. Risk Factors for Chronic Obstructive Pulmonary Disease in a European Cohort of Young Adults. Am. J. Respir. Crit. Care Med. 2012, 183, 891–897. [Google Scholar] [CrossRef] [PubMed]
  38. Stocks, J.; Sonnappa, S. Early life influences on the development of chronic obstructive pulmonary disease. Ther. Adv. Respir. Dis. 2013, 7, 161–173. [Google Scholar] [CrossRef]
  39. Savran, O.; Ulrik, C.S. Early life insults as determinants of chronic obstructive pulmonary disease in adult life. Int. J. Chron. Obstruct. Pulmon. Dis. 2018, 13, 683. [Google Scholar] [CrossRef]
  40. Shin, S.; Bai, L.; Burnett, R.T.; Kwong, J.C.; Hystad, P.; van Donkelaar, A.; Lavigne, E.; Weichenthal, S.; Copes, R.; Martin, R.V.; et al. Air pollution as a risk factor for incident chronic obstructive pulmonary disease and Asthma: A 15-year population-based cohort study. Am. J. Respir. Crit. Care Med. 2021, 203, 1138–1148. [Google Scholar] [CrossRef]
  41. Wang, L.; Xie, J.; Hu, Y.; Tian, Y. Air pollution and risk of chronic obstructed pulmonary disease: The modifying effect of genetic susceptibility and lifestyle. eBioMedicine 2022, 79, 103994. [Google Scholar] [CrossRef]
  42. Jiang, X.Q.; Mei, X.D.; Feng, D. Air pollution and chronic airway diseases: What should people know and do? J. Thorac. Dis. 2016, 8, E31. [Google Scholar] [CrossRef] [PubMed]
  43. Ali, K.M. Childhood asthma as a risk factor for adult chronic obstructive pulmonary disease: A systematic review and meta-analysis. Expert Rev. Respir. Med. 2022, 16, 461–467. [Google Scholar] [CrossRef] [PubMed]
  44. Hayden, L.P.; Cho, M.H.; Raby, B.A.; Beaty, T.H.; Silverman, E.K.; Hersh, C.P. Childhood asthma is associated with COPD and known asthma variants in COPDGene: A genome-wide association study. Respir. Res. 2018, 19, 209. [Google Scholar] [CrossRef] [PubMed]
  45. Silva, G.E.; Sherrill, D.L.; Guerra, S.; Barbee, R.A. Asthma as a Risk Factor for COPD in a Longitudinal Study. Chest 2004, 126, 59–65. [Google Scholar] [CrossRef]
  46. HANCU, A. Nutritional Status as a Risk Factor in COPD. Mædica 2019, 14, 140. [Google Scholar] [CrossRef]
  47. Rahman, I.; MacNee, W. Antioxidant pharmacological therapies for COPD. Curr. Opin. Pharmacol. 2012, 12, 256–265. [Google Scholar] [CrossRef]
  48. Hu, N.; Green, S.A. Acetyl radical generation in cigarette smoke: Quantification and simulations. Atmos. Environ. 2014, 95, 142–150. [Google Scholar] [CrossRef]
  49. Valavanidis, A.; Vlachogianni, T.; Fiotakis, K. Tobacco smoke: Involvement of reactive oxygen species and stable free radicals in mechanisms of oxidative damage, carcinogenesis and synergistic effects with other respirable particles. Int. J. Environ. Res. Public Health 2009, 6, 445–462. [Google Scholar] [CrossRef]
  50. Winterbourn, C.C. Reconciling the chemistry and biology of reactive oxygen species. Nat. Chem. Biol. 2008, 4, 278–286. [Google Scholar] [CrossRef]
  51. Yi, S.; Zhang, F.; Qu, F.; Ding, W. Water-insoluble fraction of airborne particulate matter (PM10) induces oxidative stress in human lung epithelial A549 cells. Environ. Toxicol. 2014, 29, 226–233. [Google Scholar] [CrossRef]
  52. Veal, E.A.; Day, A.M.; Morgan, B.A. Hydrogen Peroxide Sensing and Signaling. Mol. Cell 2007, 26, 1–14. [Google Scholar] [CrossRef] [PubMed]
  53. Marinho, H.S.; Real, C.; Cyrne, L.; Soares, H.; Antunes, F. Hydrogen peroxide sensing, signaling and regulation of transcription factors. Redox Biol. 2014, 2, 535–562. [Google Scholar] [CrossRef] [PubMed]
  54. Salama, S.A.; Arab, H.H.; Omar, H.A.; Maghrabi, I.A.; Snapka, R.M. Nicotine mediates hypochlorous acid-induced nuclear protein damage in mammalian cells. Inflammation 2014, 37, 785–792. [Google Scholar] [CrossRef]
  55. Hawkins, C.L.; Pattison, D.I.; Davies, M.J. Hypochlorite-induced oxidation of amino acids, peptides and proteins. Amin. Acids 2003, 25, 259–274. [Google Scholar] [CrossRef] [PubMed]
  56. Wiebel, F.J. Chemistry and Primary Toxicity of Tobacco and Tobacco Smoke. In Progress in Respiratory Research; Karger, A.G.S., Ed.; Karger AG: Basel, Switzerland, 2015; Volume 42, pp. 37–46. [Google Scholar]
  57. Thomas, D.D.; Liu, X.; Kantrow, S.P.; Jack, R.; Lancaster, J. The biological lifetime of nitric oxide: Implications for the perivascular dynamics of NO and O2. Proc. Natl. Acad. Sci. USA 2001, 98, 355. [Google Scholar] [CrossRef]
  58. Storkey, C.; Pattison, D.I.; Ignasiak, M.T.; Schiesser, C.H.; Davies, M.J. Kinetics of reaction of peroxynitrite with selenium- and sulfur-containing compounds: Absolute rate constants and assessment of biological significance. Free Radic. Biol. Med. 2015, 89, 1049–1056. [Google Scholar] [CrossRef]
  59. Goldstein, S.; Merényi, G. The Chemistry of Peroxynitrite: Implications for Biological Activity. In Methods in Enzymology; Academic Press Inc.: Cambridge, MA, USA, 2008; Volume 436, pp. 49–61. ISBN 9780123742773. [Google Scholar]
  60. Ferrer-Sueta, G.; Radi, R. Chemical biology of peroxynitrite: Kinetics, diffusion, and radicals. ACS Chem. Biol. 2009, 4, 161–177. [Google Scholar] [CrossRef]
  61. Radi, R. Peroxynitrite, a Stealthy Biological Oxidant. J. Biol. Chem. 2013, 288, 26464. [Google Scholar] [CrossRef]
  62. Fransen, M.; Nordgren, M.; Wang, B.; Apanasets, O. Role of peroxisomes in ROS/RNS-metabolism: Implications for human disease. Biochim. Biophys. Acta—Mol. Basis Dis. 2012, 1822, 1363–1373. [Google Scholar] [CrossRef]
  63. Zito, E.; Melo, E.P.; Yang, Y.; Wahlander, Å.; Neubert, T.A.; Ron, D. Oxidative protein folding by an endoplasmic reticulum localized peroxiredoxin. Mol. Cell 2010, 40, 787. [Google Scholar] [CrossRef] [Green Version]
  64. Lipinski, B. Hydroxyl radical and its scavengers in health and disease. Oxid. Med. Cell. Longev. 2011, 2011, 809696. [Google Scholar] [CrossRef] [PubMed]
  65. Domej, W.; Oettl, K.; Renner, W. Oxidative stress and free radicals in COPD-implications and relevance for treatment. Int. J. COPD 2014, 9, 1207–1224. [Google Scholar] [CrossRef] [PubMed]
  66. Wang, Y.; Branicky, R.; Noë, A.; Hekimi, S. Superoxide dismutases: Dual roles in controlling ROS damage and regulating ROS signaling. J. Cell Biol. 2018, 217, 1915–1928. [Google Scholar] [CrossRef]
  67. Tvrdá, E.; Benko, F. Free radicals: What they are and what they do. In Pathology: Oxidative Stress and Dietary Antioxidants; Elsevier: Amsterdam, The Netherlands, 2020; pp. 3–13. ISBN 9780128159729. [Google Scholar]
  68. Lennicke, C.; Rahn, J.; Lichtenfels, R.; Wessjohann, L.A.; Seliger, B. Hydrogen peroxide—Production, fate and role in redox signaling of tumor cells. Cell Commun. Signal. 2015, 13, 39. [Google Scholar] [CrossRef]
  69. Konno, T.; Melo, E.P.; Chambers, J.E.; Avezov, E. Intracellular sources of ROS/H2O2 in health and neurodegeneration: Spotlight on endoplasmic reticulum. Cells 2021, 10, 233. [Google Scholar] [CrossRef] [PubMed]
  70. Bhatt, A.; Jain, A.; Gurnany, E.; Jain, R.; Modi, A.; Jain, A. Carbon Nanotubes: A Promising Carrier for Drug Delivery and Targeting. Nanoarchitecton. Smart Deliv. Drug Target. 2016, 670815, 465–501. [Google Scholar] [CrossRef]
  71. Zhang, J.J.; Wei, Y.; Fang, Z. Ozone pollution: A major health hazard worldwide. Front. Immunol. 2019, 10, 2518. [Google Scholar] [CrossRef]
  72. Nabi, R.B.S.; Tayade, R.; Hussain, A.; Kulkarni, K.P.; Imran, Q.M.; Mun, B.G.; Yun, B.W. Nitric oxide regulates plant responses to drought, salinity, and heavy metal stress. Environ. Exp. Bot. 2019, 161, 120–133. [Google Scholar] [CrossRef]
  73. Jarvis, D.J.; Adamkiewicz, G.; Heroux, M.-E.; Rapp, R.; Kelly, F.J. Nitrogen Dioxide. In WHO Guidelines for Indoor Air Quality: Selected Pollutants; World Health Organization: Geneva, Switzerland, 2010. [Google Scholar]
  74. Rane, D.; Carlson, E.J.; Yin, Y.; Peterson, B.R. Fluorescent detection of peroxynitrite during antibody-dependent cellular phagocytosis. Methods Enzymol. 2020, 640, 1–35. [Google Scholar] [CrossRef]
  75. Zielonka, J.; Sikora, A.; Joseph, J.; Kalyanaraman, B. Peroxynitrite is the major species formed from different flux ratios of co-generated nitric oxide and superoxide: Direct reaction with boronate-based fluorescent probe. J. Biol. Chem. 2010, 285, 14210–14216. [Google Scholar] [CrossRef] [Green Version]
  76. Kassim, S.Y.; Fu, X.; Liles, W.C.; Shapiro, S.D.; Parks, W.C.; Heinecke, J.W. NADPH Oxidase Restrains the Matrix Metalloproteinase Activity of Macrophages. J. Biol. Chem. 2005, 280, 30201–30205. [Google Scholar] [CrossRef] [PubMed]
  77. Yao, H.; Edirisinghe, I.; Yang, S.R.; Rajendrasozhan, S.; Kode, A.; Caito, S.; Adenuga, D.; Rahman, I. Genetic Ablation of NADPH Oxidase Enhances Susceptibility to Cigarette Smoke-Induced Lung Inflammation and Emphysema in Mice. Am. J. Pathol. 2008, 172, 1222. [Google Scholar] [CrossRef] [PubMed]
  78. Yao, H.; Rahman, I. Current concepts on oxidative/carbonyl stress, inflammation and epigenetics in pathogenesis of chronic obstructive pulmonary disease. Toxicol. Appl. Pharmacol. 2011, 254, 72–85. [Google Scholar] [CrossRef] [PubMed]
  79. Kodgule, R.; Salvi, S. Exposure to biomass smoke as a cause for airway disease in women and children. Curr. Opin. Allergy Clin. Immunol. 2012, 12, 82–90. [Google Scholar] [CrossRef]
  80. Louhelainen, N.; Rytilä, P.; Haahtela, T.; Kinnula, V.L.; Djukanović, R. Persistence of oxidant and protease burden in the airways after smoking cessation. BMC Pulm. Med. 2009, 9, 25. [Google Scholar] [CrossRef]
  81. Van Der Toorn, M.; Rezayat, D.; Kauffman, H.F.; Bakker, S.J.L.; Gans, R.O.B.; Koëter, G.H.; Choi, A.M.K.; Van Oosterhout, A.J.M.; Slebos, D.J. Lipid-soluble components in cigarette smoke induce mitochondrial production of reactive oxygen species in lung epithelial cells. Am. J. Physiol.—Lung Cell. Mol. Physiol. 2009, 297, L109. [Google Scholar] [CrossRef]
  82. Aguilera-Aguirre, L.; Bacsi, A.; Saavedra-Molina, A.; Kurosky, A.; Sur, S.; Boldogh, I. Mitochondrial dysfunction increases allergic airway inflammation. J. Immunol. 2009, 183, 5379. [Google Scholar] [CrossRef]
  83. Aaron, S.D.; Angel, J.B.; Lunau, M.; Wright, K.; Fex, C.; Le Saux, N.; Dales, R.E. Granulocyte inflammatory markers and airway infection during acute exacerbation of chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2001, 163, 349–355. [Google Scholar] [CrossRef]
  84. Fujii, J.; Homma, T.; Osaki, T. Superoxide Radicals in the Execution of Cell Death. Antioxidants 2022, 11, 501. [Google Scholar] [CrossRef]
  85. Janssen-Heininger, Y.M.W.; Persinger, R.L.; Korn, S.H.; Pantano, C.; McElhinney, B.; Reynaert, N.L.; Langen, R.C.J.; Ckless, K.; Shrivastava, P.; Poynter, M.E. Reactive nitrogen species and cell signaling: Implications for death or survival of lung epithelium. Am. J. Respir. Crit. Care Med. 2002, 166, S9–S16. [Google Scholar] [CrossRef]
  86. Dey, T.; Kalita, J.; Weldon, S.; Taggart, C.C. Proteases and their inhibitors in chronic obstructive pulmonary disease. J. Clin. Med. 2018, 7, 244. [Google Scholar] [CrossRef] [PubMed]
  87. Janciauskiene, S.; Wrenger, S.; Immenschuh, S.; Olejnicka, B.; Greulich, T.; Welte, T.; Chorostowska-Wynimko, J. The multifaceted effects of Alpha1-Antitrypsin on neutrophil functions. Front. Pharmacol. 2018, 9, 341. [Google Scholar] [CrossRef] [PubMed]
  88. Jacob, S.; Herndon, D.N.; Hawkins, H.K.; Enkhbaatar, P.; Cox, R.A. Xanthine oxidase contributes to sustained airway epithelial oxidative stress after scald burn. Int. J. Burns Trauma 2017, 7, 98–106. [Google Scholar]
  89. Price, M.E.; Sisson, J.H. Redox regulation of motile cilia in airway disease. Redox Biol. 2019, 27, 101146. [Google Scholar] [CrossRef]
  90. Nadel, J.A. Role of epidermal growth factor receptor activation in regulating mucin synthesis. Respir. Res. 2001, 2, 85. [Google Scholar] [CrossRef]
  91. Hirst, R.A.; Sikand, K.S.; Rutman, A.; Mitchell, T.J.; Andrew, P.W.; O’Callaghan, C. Relative roles of pneumolysin and hydrogen peroxide from Streptococcus pneumoniae in inhibition of ependymal ciliary beat frequency. Infect. Immun. 2000, 68, 1557–1562. [Google Scholar] [CrossRef]
  92. Mercer, B.; Lemaitre, V.; Powell, C.; D’Armiento, J. The Epithelial Cell in Lung Health and Emphysema Pathogenesis. Curr. Respir. Med. Rev. 2006, 2, 101–142. [Google Scholar] [CrossRef]
  93. Eutamene, H.; Theodorou, V.; Schmidlin, F.; Tondereau, V.; Garcia-Villar, R.; Salvador-Cartier, C.; Chovet, M.; Bertrand, C.; Bueno, L. LPS-induced lung inflammation is linked to increased epithelial permeability: Role of MLCK. Eur. Respir. J. 2005, 25, 789–796. [Google Scholar] [CrossRef] [PubMed]
  94. Rusznak, C.; Mills, P.R.; Devalia, J.L.; Sapsford, R.J.; Davies, R.J.; Lozewicz, S. Effect of cigarette smoke on the permeability and IL-1β and sICAM-1 release from cultured human bronchial epithelial cells of never-smokers, smokers, and patients with chronic obstructive pulmonary disease. Am. J. Respir. Cell Mol. Biol. 2000, 23, 530–536. [Google Scholar] [CrossRef] [PubMed]
  95. Choudhury, G.; MacNee, W. Role of Inflammation and Oxidative Stress in the Pathology of Ageing in COPD: Potential Therapeutic Interventions. COPD J. Chronic Obstr. Pulm. Dis. 2017, 14, 122–135. [Google Scholar] [CrossRef]
  96. Gould, N.S.; Min, E.; Huang, J.; Chu, H.W.; Good, J.; Martin, R.J.; Day, B.J. Glutathione depletion accelerates cigarette smoke-induced inflammation and airspace enlargement. Toxicol. Sci. 2015, 147, 466–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Di Stefano, A.; Caramori, G.; Ricciardolo, F.L.M.; Capelli, A.; Adcock, I.M.; Donner, C.F. Cellular and molecular mechanisms in chronic obstructive pulmonary disease: An overview. Clin. Exp. Allergy 2004, 34, 1156–1167. [Google Scholar] [CrossRef] [PubMed]
  98. Rahman, I. Oxidative stress, transcription factors and chromatin remodelling in lung inflammation. Biochem. Pharmacol. 2002, 64, 935–942. [Google Scholar] [CrossRef]
  99. Di Stefano, A.; Caramori, G.; Oates, T.; Capelli, A.; Lusuardi, M.; Gnemmi, I.; Ioli, F.; Chung, K.F.; Donner, C.F.; Barnes, P.J.; et al. Increased expression of nuclear factor-κB in bronchial biopsies from smokers and patients with COPD. Eur. Respir. J. 2002, 20, 556–563. [Google Scholar] [CrossRef]
  100. MacNee, W. Oxidative stress and lung inflammation in airways disease. Eur. J. Pharmacol. 2001, 429, 195–207. [Google Scholar] [CrossRef]
  101. Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef]
  102. Morgan, M.J.; Liu, Z.G. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef]
  103. Lingappan, K. NF-κB in oxidative stress. Curr. Opin. Toxicol. 2018, 7, 81–86. [Google Scholar] [CrossRef]
  104. Parmentier, M.; Hirani, N.; Rahman, I.; Donaldson, K.; MacNee, W.; Antonicelli, F. Regulation of lipopolysaccharide-mediated interleukin-1beta release by N-acetylcysteine in THP-1 cells. Eur. Respir. J. 2000, 16, 933–939. [Google Scholar] [CrossRef]
  105. Jimenez, L.A.; Thompson, J.; Brown, D.A.; Rahman, I.; Antonicelli, F.; Duffin, R.; Drost, E.M.; Hay, R.T.; Donaldson, K.; MacNee, W. Activation of NF-κB by PM10 Occurs via an Iron-Mediated Mechanism in the Absence of IκB Degradation. Toxicol. Appl. Pharmacol. 2000, 166, 101–110. [Google Scholar] [CrossRef]
  106. Barnes, P.J.; Adcock, I.M.; Ito, K. Histone acetylation and deacetylation: Importance in inflammatory lung diseases. Eur. Respir. J. 2005, 25, 552–563. [Google Scholar] [CrossRef] [PubMed]
  107. Barnes, P.J. Role of HDAC2 in the Pathophysiology of COPD. Annu. Rev. Physiol. 2009, 71, 451–464. [Google Scholar] [CrossRef] [PubMed]
  108. Tan, Z.X.; Fu, L.; Wang, W.J.; Zhan, P.; Zhao, H.; Wang, H.; Xu, D.X. Serum CYR61 Is Associated with Airway Inflammation and Is a Potential Biomarker for Severity in Chronic Obstructive Pulmonary Disease. Front. Med. 2021, 8, 781596. [Google Scholar] [CrossRef] [PubMed]
  109. Marwick, J.A.; Kirkham, P.A.; Stevenson, C.S.; Danahay, H.; Giddings, J.; Butler, K.; Donaldson, K.; MacNee, W.; Rahman, I. Cigarette Smoke Alters Chromatin Remodeling and Induces Proinflammatory Genes in Rat Lungs. Am. J. Respir. Cell Mol. Biol. 2012, 31, 633–642. [Google Scholar] [CrossRef]
  110. Tan, B.L.; Norhaizan, M.E.; Liew, W.P.P.; Rahman, H.S. Antioxidant and oxidative stress: A mutual interplay in age-related diseases. Front. Pharmacol. 2018, 9, 1162. [Google Scholar] [CrossRef]
  111. Sharifi-Rad, M.; Anil Kumar, N.V.; Zucca, P.; Varoni, E.M.; Dini, L.; Panzarini, E.; Rajkovic, J.; Tsouh Fokou, P.V.; Azzini, E.; Peluso, I.; et al. Lifestyle, Oxidative Stress, and Antioxidants: Back and Forth in the Pathophysiology of Chronic Diseases. Front. Physiol. 2020, 11, 694. [Google Scholar] [CrossRef]
  112. Poljsak, B. Strategies for reducing or preventing the generation of oxidative stress. Oxid. Med. Cell. Longev. 2011, 2011, 194586. [Google Scholar] [CrossRef]
  113. Tuder, R.M.; Zhen, L.; Cho, C.Y.; Taraseviciene-Stewart, L.; Kasahara, Y.; Salvemini, D.; Voelkel, N.F.; Flores, S.C. 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. [Google Scholar] [CrossRef]
  114. Kasahara, Y.; Tuder, R.M.; Cool, C.D.; Lynch, D.A.; Flores, S.C.; Voelkel, N.F. 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. [Google Scholar] [CrossRef]
  115. Agustí, A.; Morlá, M.; Sauleda, J.; Saus, C.; Busquets, X. NF-κB activation and iNOS upregulation in skeletal muscle of patients with COPD and low body weight. Thorax 2004, 59, 483–487. [Google Scholar] [CrossRef]
  116. Rahman, I.; Biswas, S.K.; Jimenez, L.A.; Torres, M.; Forman, H.J. Glutathione, Stress Responses, and Redox Signaling in Lung Inflammation. Antioxid. Redox Signal. 2004, 7, 42–59. [Google Scholar] [CrossRef] [PubMed]
  117. Kode, A.; Yang, S.-R.; Rahman, I. Differential effects of cigarette smoke on oxidative stress and proinflammatory cytokine release in primary human airway epithelial cells and in a variety of transformed alveolar epithelial cells. Respir. Res. 2006, 7, 1–20. [Google Scholar] [CrossRef] [PubMed]
  118. Harju, T.; Kaarteenaho-Wiik, R.; Soini, Y.; Sormunen, R.; Kinnula, V.L. Diminished Immunoreactivity of γ-Glutamylcysteine Synthetase in the Airways of Smokers’ Lung. Am. J. Respir. Crit. Care Med. 2002, 166, 754–759. [Google Scholar] [CrossRef] [PubMed]
  119. Tomaki, M.; Sugiura, H.; Koarai, A.; Komaki, Y.; Akita, T.; Matsumoto, T.; Nakanishi, A.; Ogawa, H.; Hattori, T.; Ichinose, M. Decreased expression of antioxidant enzymes and increased expression of chemokines in COPD lung. Pulm. Pharmacol. Ther. 2007, 20, 596–605. [Google Scholar] [CrossRef]
  120. Cheng, S.L.; Yu, C.J.; Chen, C.J.; Yang, P.C. Genetic polymorphism of epoxide hydrolase and glutathione S-transferase in COPD. Eur. Respir. J. 2004, 23, 818–824. [Google Scholar] [CrossRef]
  121. DeMeo, D.L.; Hersh, C.P.; Hoffman, E.A.; Litonjua, A.A.; Lazarus, R.; Sparrow, D.; Benditt, J.O.; Criner, G.; Make, B.; Martinez, F.J.; et al. Genetic determinants of emphysema distribution in the national emphysema treatment trial. Am. J. Respir. Crit. Care Med. 2007, 176, 42–48. [Google Scholar] [CrossRef]
  122. Michaeloudes, C.; Sukkar, M.B.; Khorasani, N.M.; Bhavsar, P.K.; Chung, K.F. TGF-β regulates Nox4, MnSOD and catalase expression, and IL-6 release in airway smooth muscle cells. Am. J. Physiol.—Lung Cell. Mol. Physiol. 2011, 300, L295. [Google Scholar] [CrossRef]
  123. Hwang, J.; Rajendrasozhan, S.; Yao, H.; Chung, S.; Sundar, I.K.; Huyck, H.L.; Pryhuber, G.S.; Kinnula, V.L.; Rahman, I. FoxO3 deficiency leads to increased susceptibility to cigarette smoke-induced inflammation, airspace enlargement, and chronic obstructive pulmonary disease. J. Immunol. 2011, 187, 987. [Google Scholar] [CrossRef]
  124. Pietras, T.; Szemraj, J.; Witusik, A.; Hołub, M.; Panek, M.; Wujcik, R.; Gorski, P. The sequence polymorphism of MnSOD gene in subjects with respiratory insufficiency in COPD. Med. Sci. Monit. 2010, 16, 427–432. [Google Scholar]
  125. Dahl, M.; Bowler, R.P.; Juul, K.; Crapo, J.D.; Levy, S.; Nordestgaard, B.G. Superoxide Dismutase 3 Polymorphism Associated with Reduced Lung Function in Two Large Populations. Am. J. Respir. Crit. Care Med. 2008, 178, 906. [Google Scholar] [CrossRef]
  126. Young, R.P.; Hopkins, R.; Black, P.N.; Eddy, C.; Wu, L.; Gamble, G.D.; Mills, G.D.; Garrett, J.E.; Eaton, T.E.; Rees, M.I. Functional variants of antioxidant genes in smokers with COPD and in those with normal lung function. Thorax 2006, 61, 394. [Google Scholar] [CrossRef] [PubMed]
  127. Kobayashi, M.; Yamamoto, M. Nrf2–Keap1 regulation of cellular defense mechanisms against electrophiles and reactive oxygen species. Adv. Enzyme Regul. 2006, 46, 113–140. [Google Scholar] [CrossRef] [PubMed]
  128. Mercado, N.; Thimmulappa, R.; Thomas, C.M.R.; Fenwick, P.S.; Chana, K.K.; Donnelly, L.E.; Biswal, S.; Ito, K.; Barnes, P.J. Decreased histone deacetylase 2 impairs Nrf2 activation by oxidative stress. Biochem. Biophys. Res. Commun. 2011, 406, 292–298. [Google Scholar] [CrossRef] [PubMed]
  129. Barnes, P.J. Defective Antioxidant Gene Regulation in COPD: A Case for Broccoli. Am. J. Respir. Crit. Care Med. 2012, 178, 552–554. [Google Scholar] [CrossRef]
  130. Corradi, M.; Montuschi, P.; Donnelly, L.E.; Pesci, A.; Kharitonov, S.A.; Barnes, P.J. Increased nitrosothiols in exhaled breath condensate in inflammatory airway diseases. Am. J. Respir. Crit. Care Med. 2001, 163, 854–858. [Google Scholar] [CrossRef]
  131. Paredi, P.; Kharitonov, S.A.; Leak, D.; Ward, S.; Cramer, D.; Barnes, P.J. Exhaled ethane, a marker of lipid peroxidation, is elevated chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2000, 162, 369–373. [Google Scholar] [CrossRef]
  132. Delen, F.M.; Sippel, J.M.; Osborne, M.L.; Law, S.; Thukkani, N.; Holden, W.E. Increased exhaled nitric oxide in chronic bronchitis: Comparison with asthma and COPD. Chest 2000, 117, 695–701. [Google Scholar] [CrossRef]
  133. Griendling, K.K.; Touyz, R.M.; Zweier, J.L.; Dikalov, S.; Chilian, W.; Chen, Y.R.; Harrison, D.G.; Bhatnagar, A. Measurement of Reactive Oxygen Species, Reactive Nitrogen Species, and Redox-Dependent Signaling in the Cardiovascular System: A Scientific Statement from the American Heart Association. Circ. Res. 2016, 119, e39–e75. [Google Scholar] [CrossRef]
  134. Bryan, N.S.; Grisham, M.B. Methods to detect nitric oxide and its metabolites in biological samples. Free Radic. Biol. Med. 2007, 43, 645–657. [Google Scholar] [CrossRef]
  135. Nagababu, E.; Rifkind, J.M. Routes for Formation of S-Nitrosothiols in Blood. Cell Biochem. Biophys. 2013, 67, 385–398. [Google Scholar] [CrossRef]
  136. Keszler, A.; Zhang, Y.; Hogg, N. Reaction between nitric oxide, glutathione, and oxygen in the presence and absence of protein: How are S-nitrosothiols formed? Free Radic. Biol. Med. 2010, 48, 55–64. [Google Scholar] [CrossRef] [PubMed]
  137. Jourd’Heuil, D.; Hallén, K.; Feelisch, M.; Grisham, M.B. Dynamic state of S-nitrosothiols in human plasma and whole blood. Free Radic. Biol. Med. 2000, 28, 409–417. [Google Scholar] [CrossRef]
  138. van Beurden, W.J.C.; Dekhuijzen, P.N.R.; Harff, G.A.; Smeenk, F.W.J.M. Variability of Exhaled Hydrogen Peroxide in Stable COPD Patients and Matched Healthy Controls. Respiration 2002, 69, 211–216. [Google Scholar] [CrossRef]
  139. Barnes, P.J. Oxidative stress-based therapeutics in COPD. Redox Biol. 2020, 33, 101544. [Google Scholar] [CrossRef] [PubMed]
  140. Strzelak, A.; Ratajczak, A.; Adamiec, A.; Feleszko, W. Tobacco smoke induces and alters immune responses in the lung triggering inflammation, allergy, asthma and other lung diseases: A mechanistic review. Int. J. Environ. Res. Public Health 2018, 15, 1033. [Google Scholar] [CrossRef] [PubMed]
  141. Boots, A.W.; Haenen, G.R.M.M.; Bast, A. Oxidant metabolism in chronic obstructive pulmonary disease. Eur. Respir. J. Suppl. 2003, 22, 14s–27s. [Google Scholar] [CrossRef] [PubMed]
  142. Zhang, W.Z.; Butler, J.J.; Cloonan, S.M. Smoking-induced iron dysregulation in the lung. Free Radic. Biol. Med. 2019, 133, 238–247. [Google Scholar] [CrossRef] [PubMed]
  143. Zhao, Z. Iron and oxidizing species in oxidative stress and Alzheimer’s disease. Aging Med. 2019, 2, 82–87. [Google Scholar] [CrossRef] [PubMed]
  144. Saetta, M.; Turato, G.; Maestrelli, P.; Mapp, C.E.; Fabbri, L.M. Cellular and structural bases of chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2001, 163, 1304–1309. [Google Scholar] [CrossRef]
  145. Vassallo, R.; Kroening, P.R.; Parambil, J.; Kita, H. Nicotine and oxidative cigarette smoke constituents induce immune-modulatory and pro-inflammatory dendritic cell responses. Mol. Immunol. 2008, 45, 3321–3329. [Google Scholar] [CrossRef]
  146. Chung, K.F. Cytokines in chronic obstructive pulmonary disease. Eur. Respir. J. Suppl. 2001, 18, 50s–59s. [Google Scholar] [CrossRef]
  147. Aghasafari, P.; George, U.; Pidaparti, R. A review of inflammatory mechanism in airway diseases. Inflamm. Res. 2019, 68, 59–74. [Google Scholar] [CrossRef] [PubMed]
  148. Facchinetti, F.; Amadei, F.; Geppetti, P.; Tarantini, F.; Di Serio, C.; Dragotto, A.; Gigli, P.M.; Catinella, S.; Civelli, M.; Patacchini, R. α,β-unsaturated aldehydes in cigarette smoke release inflammatory mediators from human macrophages. Am. J. Respir. Cell Mol. Biol. 2007, 37, 617–623. [Google Scholar] [CrossRef] [PubMed]
  149. Bernard, K.; Hecker, L.; Luckhardt, T.R.; Cheng, G.; Thannickal, V.J. NADPH oxidases in lung health and disease. Antioxid. Redox Signal. 2014, 20, 2838–2853. [Google Scholar] [CrossRef] [PubMed]
  150. Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive oxygen species in inflammation and tissue injury. Antioxid. Redox Signal. 2014, 20, 1126–1167. [Google Scholar] [CrossRef]
  151. Panday, A.; Sahoo, M.K.; Osorio, D.; Batra, S. NADPH oxidases: An overview from structure to innate immunity-associated pathologies. Cell. Mol. Immunol. 2015, 12, 5–23. [Google Scholar] [CrossRef] [PubMed]
  152. Hawkins, P.; McEnery, T.; Gabillard-Lefort, C.; Bergin, D.A.; Alfawaz, B.; Shutchaidat, V.; Meleady, P.; Henry, M.; Coleman, O.; Murphy, M.; et al. In vitro and in vivo modulation of NADPH oxidase activity and reactive oxygen species production in human neutrophils by α1-antitrypsin. ERJ Open Res. 2021, 7, 00234–2021. [Google Scholar] [CrossRef] [PubMed]
  153. Kellner, M.; Noonepalle, S.; Lu, Q.; Srivastava, A.; Zemskov, E.; Black, S.M. ROS signaling in the pathogenesis of Acute Lung Injury (ALI) and Acute Respiratory Distress Syndrome (ARDS). Adv. Exp. Med. Biol. 2017, 967, 105–137. [Google Scholar] [CrossRef]
  154. O’Donnell, R.; Breen, D.; Wilson, S.; Djukanovic, R. Inflammatory cells in the airways in COPD. Thorax 2006, 61, 448–454. [Google Scholar] [CrossRef]
  155. Fujimoto, K.; Yasuo, M.; Urushibata, K.; Hanaoka, M.; Koizumi, T.; Kubo, K. Airway inflammation during stable and acutely exacerbated chronic obstructive pulmonary disease. Eur. Respir. J. 2005, 25, 640–646. [Google Scholar] [CrossRef]
  156. O’Driscoll, B.R.; Howard, L.S.; Davison, A.G. BTS guideline for emergency oxygen use in adult patients. Thorax 2008, 63, vi1–vi68. [Google Scholar] [CrossRef]
  157. Agusti, A.; Hedner, J.; Marin, J.M.; Barbéle, F.; Cazzola, M.; Rennard, S. Night-time symptoms: A forgotten dimension of COPD. Eur. Respir. Rev. 2011, 20, 183–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Butler, A.; Walton, G.M.; Sapey, E. Neutrophilic Inflammation in the Pathogenesis of Chronic Obstructive Pulmonary Disease. COPD J. Chronic Obstr. Pulm. Dis. 2018, 15, 392–404. [Google Scholar] [CrossRef]
  159. Agustí, A.G.N.; Noguera, A.; Sauleda, J.; Sala, E.; Pons, J.; Busquets, X. Systemic effects of chronic obstructive pulmonary disease. Eur. Respir. J. 2003, 21, 347–360. [Google Scholar] [CrossRef] [PubMed]
  160. Ayala, A.; Muñoz, M.F.; Argüelles, S. Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid. Med. Cell. Longev. 2014, 2014, 360438. [Google Scholar] [CrossRef]
  161. Das, K.; Roychoudhury, A. Reactive oxygen species (ROS) and response of antioxidants as ROS-scavengers during environmental stress in plants. Front. Environ. Sci. 2014, 2, 53. [Google Scholar] [CrossRef]
  162. Rahman, I.; Biswas, S.K. Non-invasive biomarkers of oxidative stress: Reproducibility and methodological issues. Redox Rep. 2004, 9, 125–143. [Google Scholar] [CrossRef] [PubMed]
  163. Tsukagoshi, H.; Shimizu, Y.; Iwamae, S.; Hisada, T.; Ishizuka, T.; Iizuka, K.; Dobashi, K.; Mori, M. Evidence of oxidative stress in asthma and COPD: Potential inhibitory effect of theophylline. Respir. Med. 2000, 94, 584–588. [Google Scholar] [CrossRef] [PubMed]
  164. Donohue, J.F. Ageing, smoking and oxidative stress. Thorax 2006, 61, 461–462. [Google Scholar] [CrossRef]
  165. Foronjy, R.; D’Armiento, J. The effect of cigarette smoke-derived oxidants on the inflammatory response of the lung. Clin. Appl. Immunol. Rev. 2006, 6, 53–72. [Google Scholar] [CrossRef]
  166. Rahman, I.; Van Schadewijk, A.A.M.; Crowther, A.J.L.; Hiemstra, P.S.; Stolk, J.; MacNee, W.; De Boer, W.I. 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. [Google Scholar] [CrossRef]
  167. Reeg, S.; Grune, T. Protein Oxidation in Aging: Does It Play a Role in Aging Progression? Antioxid. Redox Signal. 2015, 23, 239–255. [Google Scholar] [CrossRef] [Green Version]
  168. Grimm, S.; Hoehn, A.; Davies, K.J.; Grune, T. Protein oxidative modifications in the ageing brain: Consequence for the onset of neurodegenerative disease. Free Radic. Res. 2011, 45, 73–88. [Google Scholar] [CrossRef] [PubMed]
  169. Grune, T. Oxidative stress, aging and the proteasomal system. Biogerontology 2000, 1, 31–40. [Google Scholar] [CrossRef] [PubMed]
  170. Stadtman, E.R. Protein oxidation and aging. Free Radic. Res. 2006, 40, 1250–1258. [Google Scholar] [CrossRef]
  171. Zappacosta, B.; Persichilli, S.; Mordente, A.; Minucci, A.; Lazzaro, D.; Meucci, E.; Giardina, B. Inhibition of salivary enzymes by cigarette smoke and the protective role of glutathione. Human & experimental toxicology 2002, 21, 7–11. [Google Scholar] [CrossRef]
  172. Yeh, C.C.; Graham Barr, R.; Powell, C.A.; Mesia-Vela, S.; Wang, Y.; Hamade, N.K.; Austin, J.H.M.; Santella, R.M. No effect of cigarette smoking dose on oxidized plasma proteins. Environ. Res. 2008, 106, 219–225. [Google Scholar] [CrossRef]
  173. Nagler, R.; Lischinsky, S.; Diamond, E.; Drigues, N.; Klein, I.; Reznick, A.Z. Effect of Cigarette Smoke on Salivary Proteins and Enzyme Activities. Arch. Biochem. Biophys. 2000, 379, 229–236. [Google Scholar] [CrossRef]
  174. Panda, K.; Chattopadhyay, R.; Chattopadhyay, D.; Chatterjee, I.B. Cigarette smoke-induced protein oxidation and proteolysis is exclusively caused by its tar phase: Prevention by vitamin C. Toxicol. Lett. 2001, 123, 21–32. [Google Scholar] [CrossRef]
  175. Pignatelli, B.; Li, C.-Q.; Boffetta, P.; Chen, Q.; Ahrens, W.; Nyberg, F.; Mukeria, A.; Bruske-Hohlfeld, I.; Fortes, C.; Constantinescu, V.; et al. Nitrated and Oxidized Plasma Proteins in Smokers and Lung Cancer Patients1. Cancer Res. 2001, 61, 778–784. [Google Scholar]
  176. Pizzimenti, S.; Ciamporcero, E.; Daga, M.; Pettazzoni, P.; Arcaro, A.; Cetrangolo, G.; Minelli, R.; Dianzani, C.; Lepore, A.; Gentile, F.; et al. Interaction of aldehydes derived from lipid peroxidation and membrane proteins. Front. Physiol. 2013, 4, 242. [Google Scholar] [CrossRef]
  177. Lopachin, R.M.; Gavin, T. Molecular mechanisms of aldehyde toxicity: A chemical perspective. Chem. Res. Toxicol. 2014, 27, 1081–1091. [Google Scholar] [CrossRef] [PubMed]
  178. Ahsan, H. 3-Nitrotyrosine: A biomarker of nitrogen free radical species modified proteins in systemic autoimmunogenic conditions. Hum. Immunol. 2013, 74, 1392–1399. [Google Scholar] [CrossRef] [PubMed]
  179. Kumar, V.; Calamaras, T.D.; Haeussler, D.; Colucci, W.S.; Cohen, R.A.; McComb, M.E.; Pimentel, D.; Bachschmid, M.M. Cardiovascular redox and ox stress proteomics. Antioxid. Redox Signal. 2012, 17, 1528–1559. [Google Scholar] [CrossRef]
  180. Ichinose, M.; Sugiura, H.; Yamagata, S.; Koarai, A.; Shirato, K. Increase in Reactive Nitrogen Species Production in Chronic Obstructive Pulmonary Disease Airways. Am. J. Respir. Crit. Care Med. 2000, 162, 701–706. [Google Scholar] [CrossRef] [PubMed]
  181. Dunlea, D.; Fee, L.; McEnery, T.; McElvaney, N.G.; Reeves, E. The impact of alpha-1 antitrypsin augmentation therapy on neutrophil-driven respiratory disease in deficient individuals. J. Inflamm. Res. 2018, 11, 123–134. [Google Scholar] [CrossRef] [PubMed]
  182. Janciauskiene, S.M.; Bals, R.; Koczulla, R.; Vogelmeier, C.; Köhnlein, T.; Welte, T. The discovery of α1-antitrypsin and its role in health and disease. Respir. Med. 2011, 105, 1129–1139. [Google Scholar] [CrossRef] [PubMed]
  183. Stadtman, E.R.; Van Remmen, H.; Richardson, A.; Wehr, N.B.; Levine, R.L. Methionine oxidation and aging. Biochim. Biophys. Acta—Proteins Proteom. 2005, 1703, 135–140. [Google Scholar] [CrossRef]
  184. Kim, G.; Weiss, S.J.; Levine, R.L. Methionine oxidation and reduction in proteins. Biochim. Biophys. Acta—Gen. Subj. 2014, 1840, 901–905. [Google Scholar] [CrossRef]
  185. Hooper, C.; Calvert, J. The role for S-carboxymethylcysteine (carbocisteine) in the management of chronic obstructive pulmonary disease. Int. J. COPD 2008, 3, 659–669. [Google Scholar] [CrossRef]
  186. Hanaoka, M.; Droma, Y.; Chen, Y.; Agatsuma, T.; Kitaguchi, Y.; Voelkel, N.F.; Kubo, K. Carbocisteine Protects Against Emphysema Induced by Cigarette Smoke Extract in Rats. Chest 2011, 139, 1101–1108. [Google Scholar] [CrossRef] [PubMed]
  187. Carpagnano, G.E.; Resta, O.; Foschino-Barbaro, M.P.; Spanevello, A.; Stefano, A.; Di Gioia, G.; Serviddio, G.; Gramiccioni, E. Exhaled Interleukine-6 and 8-isoprostane in chronic obstructive pulmonary disease: Effect of carbocysteine lysine salt monohydrate (SCMC-Lys). Eur. J. Pharmacol. 2004, 505, 169–175. [Google Scholar] [CrossRef] [PubMed]
  188. Tatsumi, K.; Fukuchi, Y. Carbocisteine improves quality of life in patients with chronic obstructive pulmonary disease. J. Am. Geriatr. Soc. 2007, 55, 1884–1886. [Google Scholar] [CrossRef]
  189. Cakan, G.; Turkoz, M.; Turan, T.; Ahmed, K.; Nagatake, T. S-carboxymethylcysteine inhibits the attachment of Streptococcus pneumoniae to human pharyngeal epithelial cells. Microb. Pathog. 2003, 34, 261–265. [Google Scholar] [CrossRef]
  190. Passi, M.; Shahid, S.; Chockalingam, S.; Sundar, I.K.; Packirisamy, G. Conventional and nanotechnology based approaches to combat chronic obstructive pulmonary disease: Implications for chronic airway diseases. Int. J. Nanomed. 2020, 15, 3803–3826. [Google Scholar] [CrossRef] [PubMed]
  191. Macciò, A.; Madeddu, C.; Panzone, F.; Mantovani, G. Carbocysteine: Clinical experience and new perspectives in the treatment of chronic inflammatory diseases. Expert Opin. Pharmacother. 2009, 10, 693–703. [Google Scholar] [CrossRef]
  192. Zheng, J.P.; Kang, J.; Huang, S.G.; Chen, P.; Yao, W.Z.; Yang, L.; Bai, C.X.; Wang, C.Z.; Wang, C.; Chen, B.Y.; et al. Effect of carbocisteine on acute exacerbation of chronic obstructive pulmonary disease (PEACE Study): A randomised placebo-controlled study. Lancet 2008, 371, 2013–2018. [Google Scholar] [CrossRef]
  193. Allegro, L.; Cordaro, C.I.; Grassi, C. Prevention of acute exacerbations of chronic obstructive bronchitis with carbocysteine lysine salt monohydrate: A multicenter, double-blind, placebo-controlled trial. Respiration 1996, 63, 174–180. [Google Scholar] [CrossRef]
  194. Edwards, G.F.; Steel, A.E.; Scott, J.K.; Jordan, J.W. S-Carboxymethylcysteine in the fluidification of sputum and treatment of chronic airway obstruction. Chest 1976, 70, 506–513. [Google Scholar] [CrossRef]
  195. Aylward, M. An assessment of s-carboxymethylcysteine in the treatment of chronic bronchitis. Curr. Med. Res. Opin. 1974, 2, 387–394. [Google Scholar] [CrossRef]
  196. Yasuda, H.; Yamaya, M.; Sasaki, T.; Inoue, D.; Nakayama, K.; Yamada, M.; Asada, M.; Yoshida, M.; Suzuki, T.; Nishimura, H.; et al. Carbocisteine inhibits rhinovirus infection in human tracheal epithelial cells. Eur. Respir. J. 2006, 28, 51–58. [Google Scholar] [CrossRef] [PubMed]
  197. Yasuda, H.; Yamaya, M.; Sasaki, T.; Inoue, D.; Nakayama, K.; Tomita, N.; Yoshida, M.; Sasaki, H. Carbocisteine Reduces Frequency of Common Colds and Exacerbations in Patients with Chronic Obstructive Pulmonary Disease. J. Am. Geriatr. Soc. 2006, 54, 378–380. [Google Scholar] [CrossRef] [PubMed]
  198. Rubio, M.L.; Martin-Mosquero, M.C.; Ortega, M.; Peces-Barba, G.; González-Mangado, N. Oral N-Acetylcysteine Attenuates Elastase-Induced Pulmonary Emphysema in Rats. Chest 2004, 125, 1500–1506. [Google Scholar] [CrossRef] [Green Version]
  199. Alam, S.; Li, Z.; Janciauskiene, S.; Mahadeva, R. Oxidation of Z α1-Antitrypsin by Cigarette Smoke Induces Polymerization. Am. J. Respir. Cell Mol. Biol. 2011, 45, 261–269. [Google Scholar] [CrossRef]
  200. Sanguinetti, C.M. N-acetylcysteine in COPD: Why, how, and when? Multidiscip. Respir. Med. 2015, 11, 8. [Google Scholar] [CrossRef] [PubMed]
  201. Dekhuijzen, P.N.R. Antioxidant properties of N-acetylcysteine: Their relevance in relation to chronic obstructive pulmonary disease. Eur. Respir. J. 2004, 23, 629–636. [Google Scholar] [CrossRef] [PubMed]
  202. Decramer, M.; Dekhuijzen, P.N.R.; Troosters, T.; van Herwaarden, C.; Rutten-van Mölken, M.; van Schayck, C.P.O.; Olivieri, D.; Lankhorst, I.; Ardia, A. The Bronchitis Randomized on NAC Cost-Utility Study (BRONCUS): Hypothesis and design. Eur. Respir. J. 2001, 17, 329–336. [Google Scholar] [CrossRef]
  203. Decramer, M.; Rutten-van Mölken, M.; Dekhuijzen, P.N.R.; Troosters, T.; van Herwaarden, C.; Pellegrino, R.; van Schayck, C.P.O.; Olivieri, D.; Del Donno, M.; De Backer, W.; et al. Effects of N-acetylcysteine on outcomes in chronic obstructive pulmonary disease (Bronchitis Randomized on NAC Cost-Utility Study, BRONCUS): A randomised placebo-controlled trial. Lancet 2005, 365, 1552–1560. [Google Scholar] [CrossRef]
  204. Kasielski, M.; Nowak, D. Long-term administration of N-acetylcysteine decreases hydrogen peroxide exhalation in subjects with chronic obstructive pulmonary disease. Respir. Med. 2001, 95, 448–456. [Google Scholar] [CrossRef]
  205. Black, P.N.; Morgan-Day, A.; McMillan, T.E.; Poole, P.J.; Young, R.P. Randomised, controlled trial of N-acetylcysteine for treatment of acute exacerbations of chronic obstructive pulmonary disease [ISRCTN21676344]. BMC Pulm. Med. 2004, 4, 13. [Google Scholar] [CrossRef]
  206. Stey, C.; Steurer, J.; Bachmann, S.; Medici, T.C.; Tramer, M.R. The effect of oral N-acetylcysteine in chronic bronchitis: A quantitative systematic review. Eur. Respir. J. 2000, 16, 253–262. [Google Scholar] [CrossRef] [PubMed]
  207. De Benedetto, F.; Aceto, A.; Dragani, B.; Spacone, A.; Formisano, S.; Pela, R.; Donner, C.F.; Sanguinetti, C.M. Long-term oral n-acetylcysteine reduces exhaled hydrogen peroxide in stable COPD. Pulm. Pharmacol. Ther. 2005, 18, 41–47. [Google Scholar] [CrossRef] [PubMed]
  208. Poole, P.J.; Black, P.N. Oral mucolytic drugs for exacerbations of chronic obstructive pulmonary disease: Systematic review. BMJ 2001, 322, 1271–1274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Rahman, I. Antioxidant therapeutic advances in COPD. Ther. Adv. Respir. Dis. 2008, 2, 351–374. [Google Scholar] [CrossRef]
  210. Dekhuijzen, P.N.; van Beurden, W.J. The role for N-acetylcysteine in the management of COPD. Int. J. Chron. Obstruct. Pulmon. Dis. 2006, 1, 99–106. [Google Scholar] [CrossRef]
  211. Koechlin, C.; Couillard, A.; Cristol, J.P.; Chanez, P.; Hayot, M.; Le Gallais, D.; Préfaut, C. Does systemic inflammation trigger local exercise-induced oxidative stress in COPD? Eur. Respir. J. 2004, 23, 538–544. [Google Scholar] [CrossRef]
  212. Ryrsø, C.K.; Godtfredsen, N.S.; Kofod, L.M.; Lavesen, M.; Mogensen, L.; Tobberup, R.; Farver-Vestergaard, I.; Callesen, H.E.; Tendal, B.; Lange, P.; et al. Lower mortality after early supervised pulmonary rehabilitation following COPD-exacerbations: A systematic review and meta-analysis 11 Medical and Health Sciences 1103 Clinical Sciences 11 Medical and Health Sciences 1117 Public Health and Health Services 11 Medical and Health Sciences 1102 Cardiorespiratory Medicine and Haematology. BMC Pulm. Med. 2018, 18, 154. [Google Scholar] [CrossRef]
  213. Andreas, S.; Röver, C.; Heinz, J.; Straube, S.; Watz, H.; Friede, T. Decline of COPD exacerbations in clinical trials over two decades–A systematic review and meta-regression. Respir. Res. 2019, 20, 186. [Google Scholar] [CrossRef]
  214. Rhee, C.K.; Kang, C.M.; You, M.B.; Yoon, H.K.; Kim, Y.K.; Kim, K.H.; Moon, H.S.; Park, S.H.; Song, J.S. Effect of fudosteine on mucin production. Eur. Respir. J. 2008, 32, 1195–1202. [Google Scholar] [CrossRef]
  215. Osoata, G.O.; Hanazawa, T.; Brindicci, C.; Ito, M.; Barnes, P.J.; Kharitonov, S.; Ito, K. Peroxynitrite Elevation in Exhaled Breath Condensate of COPD and Its Inhibition by Fudosteine. Chest 2009, 135, 1513–1520. [Google Scholar] [CrossRef]
  216. Komatsu, H.; Yamaguchi, S.; Komorita, N.; Goto, K.; Takagi, S.; Ochi, H.; Okumoto, T. Inhibition of endotoxin- and antigen-induced airway inflammation by fudosteine, a mucoactive agent. Pulm. Pharmacol. Ther. 2005, 18, 121–127. [Google Scholar] [CrossRef] [PubMed]
  217. Moretti, M. Pharmacology and clinical efficacy of erdosteine in chronic obstructive pulmonary disease. Expert Rev. Respir. Med. 2007, 1, 307–316. [Google Scholar] [CrossRef]
  218. Moretti, M.; Bottrighi, P.; Dallari, R.; Da Porto, R.; Dolcetti, A.; Grandi, P.; Garuti, G.; Guffanti, E.; Roversi, P.; De Gugliemo, M.; et al. The effect of long-term treatment with erdosteine on chronic obstructive pulmonary disease: The EQUALIFE Study—PubMed. Drugs Exp. Clin. Res. 2004, 30, 143–152. [Google Scholar] [PubMed]
  219. Cazzola, M.; Floriani, I.; Page, C.P. The therapeutic efficacy of erdosteine in the treatment of chronic obstructive bronchitis: A meta-analysis of individual patient data. Pulm. Pharmacol. Ther. 2010, 23, 135–144. [Google Scholar] [CrossRef] [PubMed]
  220. Negro, R.W.D. Erdosteine: Antitussive and Anti-inflammatory Effects. Lung 2008, 186, 70–73. [Google Scholar] [CrossRef] [PubMed]
  221. Moretto, N.; Facchinetti, F.; Southworth, T.; Civelli, M.; Singh, D.; Patacchini, R. α,β-Unsaturated aldehydes contained in cigarette smoke elicit IL-8 release in pulmonary cells through mitogen-activated protein kinases. Am. J. Physiol.—Lung Cell. Mol. Physiol. 2009, 296, 839–848. [Google Scholar] [CrossRef]
  222. Dal Negro, R.W.; Visconti, M.; Tognella, S.; Micheletto, C. Erdosteine affects eicosanoid production in COPD. Int. J. Clin. Pharmacol. Ther. 2011, 49, 41–45. [Google Scholar] [CrossRef]
  223. Muscoli, C.; Sacco, I.; Alecce, W.; Palma, E.; Nisticò, R.; Costa, N.; Clementi, F.; Rotiroti, D.; Romeo, F.; Salvemini, D.; et al. The Protective Effect of Superoxide Dismutase Mimetic M40401 on Balloon Injury-Related Neointima Formation: Role of the Lectin-Like Oxidized Low-Density Lipoprotein Receptor-1. J. Pharmacol. Exp. Ther. 2004, 311, 44–50. [Google Scholar] [CrossRef]
  224. Sharpe, M.A.; Ollosson, R.; Stewart, V.C.; Clark, J.B. Oxidation of nitric oxide by oxomanganese-salen complexes: A new mechanism for cellular protection by superoxide dismutase/catalase mimetics. Biochem. J. 2002, 366, 97–107. [Google Scholar] [CrossRef]
  225. Smith, K.R.; Uyeminami, D.L.; Kodavanti, U.P.; Crapo, J.D.; Chang, L.-Y.; Pinkerton, K.E. Inhibition of tobacco smoke-induced lung inflammation by a catalytic antioxidant 1 11The research described in this article has been reviewed by the National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency and ap. Free Radic. Biol. Med. 2002, 33, 1106–1114. [Google Scholar] [CrossRef]
  226. Ferrer-Sueta, G.; Vitturi, D.; Batinić-Haberle, I.; Fridovich, I.; Goldstein, S.; Czapski, G.; Radi, R. Reactions of Manganese Porphyrins with Peroxynitrite and Carbonate Radical Anion *. J. Biol. Chem. 2003, 278, 27432–27438. [Google Scholar] [CrossRef] [PubMed]
  227. Tse, H.M.; Milton, M.J.; Piganelli, J.D. Mechanistic analysis of the immunomodulatory effects of a catalytic antioxidant on antigen-presenting cells: Implication for their use in targeting oxidation–reduction reactions in innate immunity. Free Radic. Biol. Med. 2004, 36, 233–247. [Google Scholar] [CrossRef] [PubMed]
  228. Vujaskovic, Z.; Batinic-Haberle, I.; Rabbani, Z.N.; Feng, Q.; Kang, S.K.; Spasojevic, I.; Samulski, T.V.; Fridovich, I.; Dewhirst, M.W.; Anscher, M.S. A small molecular weight catalytic metalloporphyrin antioxidant with superoxide dismutase (SOD) mimetic properties protects lungs from radiation-induced injury. Free Radic. Biol. Med. 2002, 33, 857–863. [Google Scholar] [CrossRef]
  229. Kinnula, V.L.; Crapo, J.D. Superoxide Dismutases in the Lung and Human Lung Diseases. Am. J. Respir. Crit. Care Med. 2003, 167, 1600–1619. [Google Scholar] [CrossRef] [PubMed]
  230. Tollefson, A.K.; Oberley-Deegan, R.E.; Butterfield, K.T.; Nicks, M.E.; Weaver, M.R.; Remigio, L.K.; Decsesznak, J.; Chu, H.W.; Bratton, D.L.; Riches, D.W.; et al. Endogenous enzymes (NOX and ECSOD) regulate smoke-induced oxidative stress. Free Radic. Biol. Med. 2010, 49, 1937–1946. [Google Scholar] [CrossRef]
  231. Yao, H.; Arunachalam, G.; Hwang, J.-W.; Chung, S.; Sundar, I.K.; Kinnula, V.L.; Crapo, J.D.; Rahman, I. Extracellular superoxide dismutase protects against pulmonary emphysema by attenuating oxidative fragmentation of ECM. Proc. Natl. Acad. Sci. USA 2010, 107, 15571–15576. [Google Scholar] [CrossRef]
  232. Kensler, T.W.; Wakabayashi, N.; Biswal, S. Cell Survival Responses to Environmental Stresses Via the Keap1-Nrf2-ARE Pathway. Annu. Rev. Pharmacol. Toxicol. 2007, 47, 89–116. [Google Scholar] [CrossRef]
  233. Rangasamy, T.; Guo, J.; Mitzner, W.A.; Roman, J.; Singh, A.; Fryer, A.D.; Yamamoto, M.; Kensler, T.W.; Tuder, R.M.; Georas, S.N.; et al. Disruption of Nrf2 enhances susceptibility to severe airway inflammation and asthma in mice. J. Exp. Med. 2005, 202, 47. [Google Scholar] [CrossRef]
  234. Sussan, T.E.; Rangasamy, T.; Blake, D.J.; Malhotra, D.; El-Haddad, H.; Bedja, D.; Yates, M.S.; Kombairaju, P.; Yamamoto, M.; Liby, K.T.; et al. Targeting Nrf2 with the triterpenoid CDDO- imidazolide attenuates cigarette smoke-induced emphysema and cardiac dysfunction in mice. Proc. Natl. Acad. Sci. USA 2009, 106, 250. [Google Scholar] [CrossRef]
  235. Rangasamy, T.; Cho, C.Y.; Thimmulappa, R.K.; Zhen, L.; Srisuma, S.S.; Kensler, T.W.; Yamamoto, M.; Petrache, I.; Tuder, R.M.; Biswal, S. Genetic ablation of Nrf2 enhances susceptibility to cigarette smoke–induced emphysema in mice. J. Clin. Invest. 2004, 114, 1248. [Google Scholar] [CrossRef]
  236. Iizuka, T.; Ishii, Y.; Itoh, K.; Kiwamoto, T.; Kimura, T.; Matsuno, Y.; Morishima, Y.; Hegab, A.E.; Homma, S.; Nomura, A.; et al. Nrf2-deficient mice are highly susceptible to cigarette smoke-induced emphysema. Genes Cells 2005, 10, 1113–1125. [Google Scholar] [CrossRef] [PubMed]
  237. Goven, D.; Boutten, A.; Leçon-Malas, V.; Marchal-Sommé, J.; Amara, N.; Crestani, B.; Foumier, M.; Lesèche, G.; Soler, P.; Boczkowski, J.; et al. Altered Nrf2/Keap1-Bach1 equilibrium in pulmonary emphysema. Thorax 2008, 63, 916–924. [Google Scholar] [CrossRef] [PubMed]
  238. Malhotra, D.; Thimmulappa, R.; Navas-Acien, A.; Sandford, A.; Elliott, M.; Singh, A.; Chen, L.; Zhuang, X.; Hogg, J.; Pare, P.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Kode, A.; Rajendrasozhan, S.; Caito, S.; Yang, S.R.; Megson, I.L.; Rahman, I. Resveratrol induces glutathione synthesis by activation of Nrf2 and protects against cigarette smoke-mediated oxidative stress in human lung epithelial cells. Am. J. Physiol.—Lung Cell. Mol. Physiol. 2008, 294, 478–488. [Google Scholar] [CrossRef]
  240. Suzuki, M.; Betsuyaku, T.; Ito, Y.; Nagai, K.; Nasuhara, Y.; Kaga, K.; Kondo, S.; Nishimura, M. Down-Regulated NF-E2–Related Factor 2 in Pulmonary Macrophages of Aged Smokers and Patients with Chronic Obstructive Pulmonary Disease. Am. J. Respir. Cell Mol. Biol. 2012, 39, 673–682. [Google Scholar] [CrossRef]
  241. Malhotra, D.; Thimmulappa, R.K.; Mercado, N.; Ito, K.; Kombairaju, P.; Kumar, S.; Ma, J.; Feller-Kopman, D.; Wise, R.; Barnes, P.; et al. Denitrosylation of HDAC2 by targeting Nrf2 restores glucocorticosteroid sensitivity in macrophages from COPD patients. J. Clin. Invest. 2011, 121, 4289. [Google Scholar] [CrossRef]
  242. Liu, Y.C.; Hsieh, C.W.; Wu, C.C.; Wung, B.S. Chalcone inhibits the activation of NF-κB and STAT3 in endothelial cells via endogenous electrophile. Life Sci. 2007, 80, 1420–1430. [Google Scholar] [CrossRef]
  243. Lee, J.H.; Haeng, S.J.; Phan, M.G.; Jin, X.; Lee, S.; Phan, T.S.; Lee, D.; Hong, Y.S.; Lee, K.; Jung, J.L. Blockade of Nuclear Factor-κB Signaling Pathway and Anti-Inflammatory Activity of Cardamomin, a Chalcone Analog from Alpinia conchigera. J. Pharmacol. Exp. Ther. 2006, 316, 271–278. [Google Scholar] [CrossRef]
  244. Foresti, R.; Hoque, M.; Monti, D.; Green, C.J.; Motterlini, R. Differential Activation of Heme Oxygenase-1 by Chalcones and Rosolic Acid in Endothelial Cells. J. Pharmacol. Exp. Ther. 2005, 312, 686–693. [Google Scholar] [CrossRef]
  245. Kumar, V.; Kumar, S.; Hassan, M.; Wu, H.; Thimmulappa, R.K.; Kumar, A.; Sharma, S.K.; Parmar, V.S.; Biswal, S.; Malhotra, S.V. Novel Chalcone Derivatives as Potent Nrf2 Activators in Mice and Human Lung Epithelial Cells. J. Med. Chem. 2011, 54, 4147. [Google Scholar] [CrossRef]
  246. Sies, H.; Berndt, C.; Jones, D.P. Oxidative Stress. Annu. Rev. Biochem. 2017, 86, 715–748. [Google Scholar] [CrossRef] [PubMed]
  247. Cifuentes-Pagano, E.; Csanyi, G.; Pagano, P.J. NADPH oxidase inhibitors: A decade of discovery from Nox2ds to HTS. Cell. Mol. Life Sci. 2012, 69, 2315–2325. [Google Scholar] [CrossRef] [PubMed]
  248. Altenhöfer, S.; Radermacher, K.A.; Kleikers, P.W.M.; Wingler, K.; Schmidt, H.H.H.W. Evolution of NADPH oxidase inhibitors: Selectivity and mechanisms for target engagement. Antioxid. Redox Signal. 2015, 23, 406–427. [Google Scholar] [CrossRef] [PubMed]
  249. Oostwoud, L.C.; Gunasinghe, P.; Seow, H.J.; Ye, J.M.; Selemidis, S.; Bozinovski, S.; Vlahos, R. Apocynin and ebselen reduce influenza A virus-induced lung inflammation in cigarette smoke-exposed mice. Sci. Rep. 2016, 6, 20983. [Google Scholar] [CrossRef]
  250. Stefanska, J.; Sarniak, A.; Wlodarczyk, A.; Sokolowska, M.; Doniec, Z.; Bialasiewicz, P.; Nowak, D.; Pawliczak, R. Hydrogen peroxide and nitrite reduction in exhaled breath condensate of COPD patients. Pulm. Pharmacol. Ther. 2012, 25, 343–348. [Google Scholar] [CrossRef] [PubMed]
  251. Cui, Y.; Wang, Y.; Li, G.; Ma, W.; Zhou, X.S.; Wang, J.; Liu, B. The Nox1/Nox4 inhibitor attenuates acute lung injury induced by ischemia-reperfusion in mice. PLoS ONE 2018, 13, e0209444. [Google Scholar] [CrossRef]
  252. Day, B.J. Catalytic antioxidants: A radical approach to new therapeutics. Drug Discov. Today 2004, 9, 557–566. [Google Scholar] [CrossRef]
  253. Vlahos, R.; Bozinovski, S. Glutathione peroxidase-1 as a novel therapeutic target for COPD. Redox Rep. 2013, 18, 142. [Google Scholar] [CrossRef]
  254. Geraghty, P.; Hardigan, A.A.; Wallace, A.M.; Mirochnitchenko, O.; Thankachen, J.; Arellanos, L.; Thompson, V.; D’Armiento, J.M.; Foronjy, R.F. The glutathione peroxidase 1-protein tyrosine phosphatase 1B-protein phosphatase 2A axis: A key determinant of airway inflammation and alveolar destruction. Am. J. Respir. Cell Mol. Biol. 2013, 49, 721–730. [Google Scholar] [CrossRef]
  255. József, L.; Filep, J.G. Selenium-containing compounds attenuate peroxynitrite-mediated NF-κB and AP-1 activation and interleukin-8 gene and protein expression in human leukocytes. Free Radic. Biol. Med. 2003, 35, 1018–1027. [Google Scholar] [CrossRef]
  256. Ishii, Y.; Hashimoto, K.; Hirano, K.; Morishima, Y.; Mochizuki, M.; Masuyama, K.; Nomura, A.; Sakamoto, T.; Uchida, Y.; Sagai, M.; et al. Ebselen decreases ozone-induced pulmonary inflammation in rats. Lung 2000, 178, 225–234. [Google Scholar] [CrossRef] [PubMed]
  257. Valença, S.S.; Rueff-Barroso, C.R.; Pimenta, W.A.; Melo, A.C.; Nesi, R.T.; Silva, M.A.S.; Porto, L.C. l-NAME and l-arginine differentially ameliorate cigarette smoke-induced emphysema in mice. Pulm. Pharmacol. Ther. 2011, 24, 587–594. [Google Scholar] [CrossRef] [PubMed]
  258. Brindicci, C.; Kharitonov, S.A.; Ito, M.; Elliott, M.W.; Hogg, J.C.; Barnes, P.J.; Ito, K. Nitric Oxide Synthase Isoenzyme Expression and Activity in Peripheral Lung Tissue of Patients with Chronic Obstructive Pulmonary Disease. Am. J. Respir. Crit. Care Med. 2010, 181, 21–30. [Google Scholar] [CrossRef] [Green Version]
  259. Barnes, P.J. New treatments for COPD. Nat. Rev. Drug Discov. 2002, 1, 437–446. [Google Scholar] [CrossRef]
  260. Rahman, I. Antioxidant therapies in COPD. Int. J. Chron. Obstruct. Pulmon. Dis. 2006, 1, 15–29. [Google Scholar] [CrossRef] [PubMed]
  261. Rahman, I. Pharmacological antioxidant strategies as therapeutic interventions for COPD. Biochim. Biophys. Acta—Mol. Basis Dis. 2012, 1822, 714–728. [Google Scholar] [CrossRef] [PubMed]
  262. Deletraz, A.; Tuccio, B.; Roussel, J.; Combes, M.; Cohen-Solal, C.; Fabre, P.L.; Trouillas, P.; Vignes, M.; Callizot, N.; Durand, G. Para-Substituted α-Phenyl-N- tert-butyl Nitrones: Spin-Trapping, Redox and Neuroprotective Properties. ACS Omega 2020, 5, 30989–30999. [Google Scholar] [CrossRef]
  263. Shi, H.; Timmins, G.; Monske, M.; Burdick, A.; Kalyanaraman, B.; Liu, Y.; Clément, J.-L.; Burchiel, S.; Liu, K.J. Evaluation of spin trapping agents and trapping conditions for detection of cell-generated reactive oxygen species. Arch. Biochem. Biophys. 2005, 437, 59–68. [Google Scholar] [CrossRef]
  264. Bottle, S.E.; Micallef, A.S. Synthesis and EPR spin trapping properties of a new isoindole-based nitrone: 1,1,3-trimethylisoindole N-oxide (TMINO). Org. Biomol. Chem. 2003, 1, 2581–2584. [Google Scholar] [CrossRef]
  265. Becker, D.A.; Ley, J.J.; Echegoyen, L.; Alvarado, R. Stilbazulenyl Nitrone (STAZN):  A Nitronyl-Substituted Hydrocarbon with the Potency of Classical Phenolic Chain-Breaking Antioxidants. J. Am. Chem. Soc. 2002, 124, 4678–4684. [Google Scholar] [CrossRef]
  266. Lapchak, P.A.; Chapman, D.F.; Zivin, J.A. Pharmacological Effects of the Spin Trap Agents N-t-Butyl-Phenylnitrone (PBN) and 2,2,6,6-Tetramethylpiperidine-N-Oxyl (TEMPO) in a Rabbit Thromboembolic Stroke Model. Stroke 2001, 32, 147–153. [Google Scholar] [CrossRef]
  267. Durand, G.; Prosak, R.A.; Han, Y.; Ortial, S.; Rockenbauer, A.; Pucci, B.; Villamena, F.A. Spin trapping and cytoprotective properties of fluorinated amphiphilic carrier conjugates of cyclic versus linear nitrones. Chem. Res. Toxicol. 2009, 22, 1570–1581. [Google Scholar] [CrossRef] [PubMed]
  268. Maples, K.R.; Green, A.R.; Floyd, R.A. Nitrone-related therapeutics: Potential of NXY-059 for the treatment of acute ischaemic stroke. CNS Drugs 2004, 18, 1071–1084. [Google Scholar] [CrossRef] [PubMed]
  269. Sugiura, H.; Ichinose, M.; Tomaki, M.; Ogawa, H.; Koarai, A.; Kitamuro, T.; Komaki, Y.; Akita, T.; Nishino, H.; Okamoto, S.; et al. Quantitative assessment of protein-bound tyrosine nitration in airway secretions from patients with inflammatory airway disease. Free Radic. Res. 2004, 38, 49–57. [Google Scholar] [CrossRef] [PubMed]
  270. Ricciardolo, F.L.M.; Caramori, G.; Ito, K.; Capelli, A.; Brun, P.; Abatangelo, G.; Papi, A.; Chung, K.F.; Adcock, I.; Barnes, P.J.; et al. Nitrosative stress in the bronchial mucosa of severe chronic obstructive pulmonary disease. J. Allergy Clin. Immunol. 2005, 116, 1028–1035. [Google Scholar] [CrossRef]
  271. Seimetz, M.; Parajuli, N.; Pichl, A.; Veit, F.; Kwapiszewska, G.; Weisel, F.C.; Milger, K.; Egemnazarov, B.; Turowska, A.; Fuchs, B.; et al. Inducible NOS inhibition reverses tobacco-smoke-induced emphysema and pulmonary hypertension in mice. Cell 2011, 147, 293–305. [Google Scholar] [CrossRef] [PubMed]
  272. Hansel, T.T.; Kharitonov, S.A.; Donnelly, L.E.; Erin, E.M.; Currie, M.G.; Moore, W.M.; Manning, P.T.; Recker, D.P.; Barnes, P.J. A selective inhibitor of inducible nitric oxide synthase inhibits exhaled breath nitric oxide in healthy volunteers and asthmatics. FASEB J. 2003, 17, 1298–1300. [Google Scholar] [CrossRef]
  273. Hall, E.D.; Springer, J.E. Neuroprotection and Acute Spinal Cord Injury: A Reappraisal. NeuroRx 2004, 1, 80–100. [Google Scholar] [CrossRef]
  274. Pascolo, L.; Zabucchi, G.; Gianoncelli, A.; Kourousias, G.; Trevisan, E.; Pascotto, E.; Casarsa, C.; Ryan, C.; Lucattelli, M.; Lungarella, G.; et al. Synchrotron X-ray microscopy reveals early calcium and iron interaction with crocidolite fibers in the lung of exposed mice. Toxicol. Lett. 2016, 241, 111–120. [Google Scholar] [CrossRef]
  275. Dianat, M.; Radan, M.; Badavi, M.; Mard, S.A.; Bayati, V.; Ahmadizadeh, M. Crocin attenuates cigarette smoke-induced lung injury and cardiac dysfunction by anti-oxidative effects: The role of Nrf2 antioxidant system in preventing oxidative stress. Respir. Res. 2018, 19, 58. [Google Scholar] [CrossRef]
  276. Kurt, A.; Tumkaya, L.; Turut, H.; Cure, M.C.; Cure, E.; Kalkan, Y.; Sehitoglu, I.; Acipayam, A. Protective Effects of Infliximab on Lung Injury Induced by Methotrexate. Arch. Bronconeumol. 2015, 51, 551–557. [Google Scholar] [CrossRef] [PubMed]
  277. Bulger, E.M.; Maier, R.V. Antioxidants in critical illness. Arch. Surg. 2001, 136, 1201–1207. [Google Scholar] [CrossRef] [PubMed]
  278. Tajima, S.; Bando, M.; Ishii, Y.; Hosono, T.; Yamasawa, H.; Ohno, S.; Takada, T.; Suzuki, E.; Gejyo, F.; Sugiyama, Y. Effects of edaravone, a free-radical scavenger, on bleomycin-induced lung injury in mice. Eur. Respir. J. 2008, 32, 1337–1343. [Google Scholar] [CrossRef]
  279. Kikuchi, K.; Uchikado, H.; Miyagi, N.; Morimoto, Y.; Takashi, I.; Tancharoen, S.; Miura, N.; Miyata, K.; Sakamoto, R.; Kikuchi, C.; et al. Beyond neurological disease: New targets for edaravone (Review). Int. J. Mol. Med. 2011, 28, 899–906. [Google Scholar] [CrossRef] [PubMed]
  280. Aldini, G.; Vistoli, G.; Regazzoni, L.; Benfatto, M.C.; Bettinelli, I.; Carini, M. Edaravone Inhibits Protein Carbonylation by a Direct Carbonyl-Scavenging Mechanism: Focus on Reactivity, Selectivity, and Reaction Mechanisms. Antioxid. Redox Signal. 2010, 12, 381–392. [Google Scholar] [CrossRef]
  281. Aldini, G.; Dalle-Donne, I.; Facino, R.M.; Milzani, A.; Carini, M. Intervention strategies to inhibit protein carbonylation by lipoxidation-derived reactive carbonyls. Med. Res. Rev. 2007, 27, 817–868. [Google Scholar] [CrossRef]
  282. Ito, K.; Ozasa, H.; Horikawa, S. Edaravone protects against lung injury induced by intestinal ischemia/reperfusion in rat. Free Radic. Biol. Med. 2005, 38, 369–374. [Google Scholar] [CrossRef]
  283. Zhou, J.; Wang, C.; Wu, J.; Fukunaga, A.; Cheng, Z.; Wang, J.; Yamauchi, A.; Yodoi, J.; Tian, H. Anti-Allergic and Anti-Inflammatory Effects and Molecular Mechanisms of Thioredoxin on Respiratory System Diseases. Antioxid. Redox Signal. 2020, 32, 785–801. [Google Scholar] [CrossRef]
  284. Tanabe, N.; Hoshino, Y.; Marumo, S.; Kiyokawa, H.; Sato, S.; Kinose, D.; Uno, K.; Muro, S.; Hirai, T.; Yodoi, J.; et al. Thioredoxin-1 protects against neutrophilic inflammation and emphysema progression in a mouse model of chronic obstructive pulmonary disease exacerbation. PLoS ONE 2013, 8, e79016. [Google Scholar] [CrossRef]
  285. Li, Y.; Liu, W.; Xing, G.; Tian, C.; Zhu, Y.; He, F. Direct association of hepatopoietin with thioredoxin constitutes a redox signal transduction in activation of AP-1/NF-κB. Cell. Signal. 2005, 17, 985–996. [Google Scholar] [CrossRef]
  286. Hopkins, B.L.; Neumann, C.A. Redoxins as gatekeepers of the transcriptional oxidative stress response. Redox Biol. 2019, 21, 101104. [Google Scholar] [CrossRef] [PubMed]
  287. Forrester, S.J.; Kikuchi, D.S.; Hernandes, M.S.; Xu, Q.; Griendling, K.K. Reactive oxygen species in metabolic and inflammatory signaling. Circ. Res. 2018, 122, 877–902. [Google Scholar] [CrossRef] [PubMed]
  288. Saha, S.; Buttari, B.; Panieri, E.; Profumo, E.; Saso, L. An Overview of Nrf2 Signaling Pathway and Its Role in Inflammation. Molecules 2020, 25, 5474. [Google Scholar] [CrossRef]
  289. Souza, D.G.; Vieira, A.T.; Pinho, V.; Sousa, L.P.; Andrade, A.A.; Bonjardim, C.A.; McMillan, M.; Kahn, M.; Teixeira, M.M. NF-kappaB plays a major role during the systemic and local acute inflammatory response following intestinal reperfusion injury. Br. J. Pharmacol. 2005, 145, 246–254. [Google Scholar] [CrossRef]
  290. Bachnoff, N.; Trus, M.; Atlas, D. Alleviation of oxidative stress by potent and selective thioredoxin-mimetic peptides. Free Radic. Biol. Med. 2011, 50, 1355–1367. [Google Scholar] [CrossRef]
  291. Sato, A.; Hoshino, Y.; Hara, T.; Muro, S.; Nakamura, H.; Mishima, M.; Yodoi, J. Thioredoxin-1 Ameliorates Cigarette Smoke-Induced Lung Inflammation and Emphysema in Mice. J. Pharmacol. Exp. Ther. 2008, 325, 380–388. [Google Scholar] [CrossRef]
  292. Kayyali, U.S.; Budhiraja, R.; Pennella, C.M.; Cooray, S.; Lanzillo, J.J.; Chalkley, R.; Hassoun, P.M. Upregulation of xanthine oxidase by tobacco smoke condensate in pulmonary endothelial cells. Toxicol. Appl. Pharmacol. 2003, 188, 59–68. [Google Scholar] [CrossRef]
  293. Komaki, Y.; Sugiura, H.; Koarai, A.; Tomaki, M.; Ogawa, H.; Akita, T.; Hattori, T.; Ichinose, M. Cytokine-mediated xanthine oxidase upregulation in chronic obstructive pulmonary disease’s airways. Pulm. Pharmacol. Ther. 2005, 18, 297–302. [Google Scholar] [CrossRef]
  294. Ichinose, M.; Sugiura, H.; Yamagata, S.; Koarai, A.; Tomaki, M.; Ogawa, H.; Komaki, Y.; Barnes, P.J.; Shirato, K.; Hattori, T. Xanthine oxidase inhibition reduces reactive nitrogen species production in COPD airways. Eur. Respir. J. 2003, 22, 457–461. [Google Scholar] [CrossRef]
  295. Scoditti, E.; Massaro, M.; Garbarino, S.; Toraldo, D.M. Role of Diet in Chronic Obstructive Pulmonary Disease Prevention and Treatment. Nutrients 2019, 11, 1357. [Google Scholar] [CrossRef]
  296. Biswas, S.; Hwang, J.W.; Kirkham, P.A.; Rahman, I. Pharmacological and Dietary Antioxidant Therapies for Chronic Obstructive Pulmonary Disease. Curr. Med. Chem. 2013, 20, 1496–1530. [Google Scholar] [CrossRef] [PubMed]
  297. Tsiligianni, I.G.; van der Molen, T. A systematic review of the role of vitamin insufficiencies and supplementation in COPD. Respir. Res. 2010, 11, 171. [Google Scholar] [CrossRef] [PubMed]
  298. Fischer, A.; Johansson, I.; Blomberg, A.; Sundström, B. Adherence to a Mediterranean-like Diet as a Protective Factor Against COPD: A Nested Case-Control Study. COPD J. Chronic Obstr. Pulm. Dis. 2019, 16, 272–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  299. Culpitt, S.V.; Rogers, D.F.; Fenwick, P.S.; Shah, P.; De Matos, C.; Russell, R.E.K.; Barnes, P.J.; Donnelly, L.E. Inhibition by red wine extract, resveratrol, of cytokine release by alveolar macrophages in COPD. Thorax 2003, 58, 942. [Google Scholar] [CrossRef] [PubMed]
  300. Birrell, M.A.; McCluskie, K.; Wong, S.; Donnelly, L.E.; Barnes, P.J.; Belvisi, M.G. Resveratrol, an extract of red wine, inhibits lipopolysaccharide induced airway neutrophilia and inflammatory mediators through an NF-κB-independent mechanism. FASEB J. 2005, 19, 1–22. [Google Scholar] [CrossRef]
  301. Navarro, S.; Reddy, R.; Lee, J.; Warburton, D.; Driscoll, B. Inhaled resveratrol treatments slow ageing-related degenerative changes in mouse lung. Thorax 2017, 72, 451–459. [Google Scholar] [CrossRef]
  302. Bartholome, A.; Kampkötter, A.; Tanner, S.; Sies, H.; Klotz, L.O. Epigallocatechin gallate-induced modulation of FoxO signaling in mammalian cells and C. elegans: FoxO stimulation is masked via PI3K/Akt activation by hydrogen peroxide formed in cell culture. Arch. Biochem. Biophys. 2010, 501, 58–64. [Google Scholar] [CrossRef]
  303. Zhu, A.; Ge, D.; Zhang, J.; Teng, Y.; Yuan, C.; Huang, M.; Adcock, I.M.; Barnes, P.J.; Yao, X. Sputum myeloperoxidase in chronic obstructive pulmonary disease. Eur. J. Med. Res. 2014, 19, 12. [Google Scholar] [CrossRef]
  304. Lazarevic-Pasti, T.; Leskovac, A.; Vasic, V. Myeloperoxidase Inhibitors as Potential Drugs. Curr. Drug Metab. 2015, 16, 168–190. [Google Scholar] [CrossRef]
  305. Churg, A.; Marshall, C.V.; Sin, D.D.; Bolton, S.; Zhou, S.; Thain, K.; Cadogan, E.B.; Maltby, J.; Soars, M.G.; Mallinder, P.R.; et al. Late intervention with a myeloperoxidase inhibitor stops progression of experimental chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2012, 185, 34–43. [Google Scholar] [CrossRef]
  306. Birch, J.; Barnes, P.J.; Passos, J.F. Mitochondria, telomeres and cell senescence: Implications for lung ageing and disease. Pharmacol. Ther. 2018, 183, 34–49. [Google Scholar] [CrossRef] [PubMed]
  307. Cloonan, S.M.; Choi, A.M.K. Mitochondria in lung disease. J. Clin. Invest. 2016, 126, 809–820. [Google Scholar] [CrossRef] [PubMed]
  308. Wiegman, C.H.; Michaeloudes, C.; Haji, G.; Narang, P.; Clarke, C.J.; Russell, K.E.; Bao, W.; Pavlidis, S.; Barnes, P.J.; Kanerva, J.; et al. Oxidative stress-induced mitochondrial dysfunction drives inflammation and airway smooth muscle remodeling in patients with chronic obstructive pulmonary disease. J. Allergy Clin. Immunol. 2015, 136, 769–780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  309. Belchamber, K.B.R.; Singh, R.; Batista, C.M.; Whyte, M.K.; Dockrell, D.H.; Kilty, I.; Robinson, M.J.; Wedzicha, J.A.; Barnes, P.J.; Donnelly, L.E. Defective bacterial phagocytosis is associated with dysfunctional mitochondria in COPD macrophages. Eur. Respir. J. 2019, 54, 1802244. [Google Scholar] [CrossRef]
  310. Sureshbabu, A.; Bhandari, V. Targeting mitochondrial dysfunction in lung diseases: Emphasis on mitophagy. Front. Physiol. 2013, 4, 384. [Google Scholar] [CrossRef]
  311. Murphy, M.P. Antioxidants as therapies: Can we improve on nature? Free Radic. Biol. Med. 2014, 66, 20–23. [Google Scholar] [CrossRef] [PubMed]
  312. Zinovkin, R.A.; Zamyatnin, A.A. Mitochondria-Targeted Drugs. Curr. Mol. Pharmacol. 2018, 12, 202–214. [Google Scholar] [CrossRef]
  313. Kolosova, N.G.; Stefanova, N.A.; Muraleva, N.A.; Skulachev, V.P. The mitochondria-targeted antioxidant sKQ1 but not n-acetylcysteine reverses aging-related biomarkers in rats. Aging 2012, 4, 686–694. [Google Scholar] [CrossRef]
  314. Hara, H.; Araya, J.; Ito, S.; Kobayashi, K.; Takasaka, N.; Yoshii, Y.; Wakui, H.; Kojima, J.; Shimizu, K.; Numata, T.; et al. Mitochondrial fragmentation in cigarette smoke-induced bronchial epithelial cell senescence. Am. J. Physiol. Cell. Mol. Physiol. 2013, 305, L737–L746. [Google Scholar] [CrossRef]
  315. Foronjy, R.; Wallace, A.; D’Armiento, J. The pharmokinetic limitations of antioxidant treatment for COPD. Pulm. Pharmacol. Ther. 2008, 21, 370–379. [Google Scholar] [CrossRef]
  316. Barnes, P.J. Oxidative Stress in Chronic Obstructive Pulmonary Disease. Antioxidants 2022, 11, 965. [Google Scholar] [CrossRef] [PubMed]
  317. Kurutas, E.B. The importance of antioxidants which play the role in cellular response against oxidative/nitrosative stress: Current state. Nutr. J. 2016, 15, 71. [Google Scholar] [CrossRef] [PubMed]
  318. Hunyadi, A. The mechanism(s) of action of antioxidants: From scavenging reactive oxygen/nitrogen species to redox signaling and the generation of bioactive secondary metabolites. Med. Res. Rev. 2019, 39, 2505–2533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  319. Matejovic, M.; Krouzecky, A.; Martinkova, V.; Rokyta, R.; Radej, J.; Kralova, H.; Treska, V.; Radermacher, P.; Novak, I. Effects of tempol, a free radical scavenger, on long-term hyperdynamic porcine bacteremia. Crit. Care Med. 2005, 33, 1057–1063. [Google Scholar] [CrossRef]
  320. Zhu, M.; Chen, Y.; Li, R.C. Oral absorption and bioavailability of tea catechins. Planta Med. 2000, 66, 444–447. [Google Scholar] [CrossRef]
  321. Shen, A.M.; Minko, T. Pharmacokinetics of inhaled nanotherapeutics for pulmonary delivery. J. Control. Release 2020, 326, 222–244. [Google Scholar] [CrossRef]
  322. Clark, A.R. Half a Century of Technological Advances in Pulmonary Drug Delivery: A Personal Perspective. Front. Drug Deliv. 2022, 2, 6. [Google Scholar] [CrossRef]
  323. Chen, Y.B.; Zhang, Y.B.; Wang, Y.L.; Kaur, P.; Yang, B.G.; Zhu, Y.; Ye, L.; Cui, Y.L. A novel inhalable quercetin-alginate nanogel as a promising therapy for acute lung injury. J. Nanobiotechnol. 2022, 20, 272. [Google Scholar] [CrossRef]
  324. Guo, Y.; Bera, H.; Shi, C.; Zhang, L.; Cun, D.; Yang, M. Pharmaceutical strategies to extend pulmonary exposure of inhaled medicines. Acta Pharm. Sin. B 2021, 11, 2565–2584. [Google Scholar] [CrossRef]
  325. Varesi, A.; Chirumbolo, S.; Campagnoli, L.I.M.; Pierella, E.; Piccini, G.B.; Carrara, A.; Ricevuti, G.; Scassellati, C.; Bonvicini, C.; Pascale, A. The Role of Antioxidants in the Interplay between Oxidative Stress and Senescence. Antioxidants 2022, 11, 1224. [Google Scholar] [CrossRef]
  326. Vona, R.; Pallotta, L.; Cappelletti, M.; Severi, C.; Matarrese, P. The impact of oxidative stress in human pathology: Focus on gastrointestinal disorders. Antioxidants 2021, 10, 201. [Google Scholar] [CrossRef] [PubMed]
  327. Estornut, C.; Milara, J.; Bayarri, M.A.; Belhadj, N.; Cortijo, J. Targeting Oxidative Stress as a Therapeutic Approach for Idiopathic Pulmonary Fibrosis. Front. Pharmacol. 2022, 12, 794997. [Google Scholar] [CrossRef]
  328. Deloconstantinos, G.; Villiotou, V. Gas phase oxidants of cigarette smoke increase nitric oxide synthase and xanthine oxidase activities of rabbit brain synaptosomes. Neurochem. Res. 2000, 25, 769–774. [Google Scholar] [CrossRef] [PubMed]
  329. Campbell, N.K.; Fitzgerald, H.K.; Dunne, A. Regulation of inflammation by the antioxidant haem oxygenase 1. Nature Reviews Immunology 2021, 21, 411–425. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A schematic presentation of the role of oxidative stress in the development of COPD.
Figure 1. A schematic presentation of the role of oxidative stress in the development of COPD.
Molecules 27 05542 g001
Figure 2. The effects of cigarette smoking on oxidative stress and inflammation in alveolar epithelial cells. Abbreviations: ERK—extracellular signal-regulated kinase; IL-1—interleukin 1; IL-1β—interleukin-1β; IL-6—interleukin 6; IL-8—interleukin 8; JNK—jun-N-terminal kinase; NF-κB,—nuclear factor-κB; TNF-α—tumor necrosis factor α.
Figure 2. The effects of cigarette smoking on oxidative stress and inflammation in alveolar epithelial cells. Abbreviations: ERK—extracellular signal-regulated kinase; IL-1—interleukin 1; IL-1β—interleukin-1β; IL-6—interleukin 6; IL-8—interleukin 8; JNK—jun-N-terminal kinase; NF-κB,—nuclear factor-κB; TNF-α—tumor necrosis factor α.
Molecules 27 05542 g002
Figure 3. The chemical structures of various antioxidants that can be used to target oxidative stress in the treatment of COPD.
Figure 3. The chemical structures of various antioxidants that can be used to target oxidative stress in the treatment of COPD.
Molecules 27 05542 g003
Table 1. The risk factors associated with the development of chronic obstructive pulmonary disease.
Table 1. The risk factors associated with the development of chronic obstructive pulmonary disease.
Risk FactorsReferences
ExternalSmoking[19,20]
Biomass smoke exposure[21,22]
Low socioeconomic status[23,24,25]
Occupational exposures[26,27]
InternalAlpha-1-antitrypsin deficiency[28,29]
Gender differences[30,31,32,33]
Airway mucus hypersecretion[34,35]
OtherAirway hyperresponsiveness[36,37]
Early life insults[38,39]
Air pollution[40,41,42]
Asthma[43,44,45]
Malnutrition[46]
Table 3. The clinical studies on the efficacy of carbocysteine in COPD.
Table 3. The clinical studies on the efficacy of carbocysteine in COPD.
AntioxidantStudy DesignDurationStudy OutcomesReferences
CarbocysteineDouble-blind, randomized, placebo-controlled study12 monthsProlonged (12 months) treatment with carbocysteine decreased the exacerbations in COPD patients, reduced exacerbations, no loss of lung activity, ameliorated health-related quality of life.[187,192]
CarbocysteineMulticenter, placebo-controlled, double-blind, parallel group trial6 monthsDuration of the acute respiratory illness was markedly decreased and this was linked with a marked decrease in the administration of antibiotics during the trial period, no serious adverse effects were observed.[193]
CarbocysteineRandomized controlled trial12 monthsConsistently decreased the frequency of exacerbations, did not alter the lung function.[188]
CarbocysteineDouble-blind controlled study3 monthsImproved the capacity to cough up bronchial secretions, markedly elevated the sputum volume output, ameliorated ventilation.[194]
CarbocysteineSingle-blind study8 weeksGreatly eased expectoration, increased the level of expectorated sputum, markedly increased peak expiratory flow rate, ameliorated the severity of dyspnea.[195]
CarbocysteineRandomized controlled trial12 monthsMarkedly decreased the exacerbation rate and commoncolds, no substantial differences in the extent of COPD.[196,197]
Table 4. The clinical studies on the efficacy of N-acetyl-L-cysteine in COPD.
Table 4. The clinical studies on the efficacy of N-acetyl-L-cysteine in COPD.
AntioxidantStudy DesignDurationStudy OutcomesReferences
N-acetyl-L-cysteine (NAC)Randomized, placebo-controlled trial3 yearsNAC is not effective at preventing deterioration of lung activity and exacerbations in COPD patients.[202,203]
NACDouble-blind, double dummy, randomized comparison study12
months
Long-term oral administration of NAC reduces H2O2 generation in the airways of patients with COPD.[204]
NACDouble-blind, randomized, placebo controlled trial7 daysNAC introduction in the treatment with bronchodilators and corticosteroids does not alter the outcome in acute exacerbation of COPD.[205]
NACRandomized, controlled trial 12–24 weeksPrevention of exacerbation and improved symptoms as compared to 34.6% of participants receiving a placebo.[206]
NACSingle-blinded, randomized trial2 monthsOral administration of NAC for 2 months quickly decreases the oxidative stress in the airways of patients with COPD.[207]
NACRandomized, controlled trial 2 monthsNo significant alteration in lung activity was observed; marked decrease in the duration of disability and a 29% decrease in exacerbations.[208]
NAC-5 daysIncreased levels of glutathione and cysteine on day 5.[209]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dailah, H.G. Therapeutic Potential of Small Molecules Targeting Oxidative Stress in the Treatment of Chronic Obstructive Pulmonary Disease (COPD): A Comprehensive Review. Molecules 2022, 27, 5542. https://doi.org/10.3390/molecules27175542

AMA Style

Dailah HG. Therapeutic Potential of Small Molecules Targeting Oxidative Stress in the Treatment of Chronic Obstructive Pulmonary Disease (COPD): A Comprehensive Review. Molecules. 2022; 27(17):5542. https://doi.org/10.3390/molecules27175542

Chicago/Turabian Style

Dailah, Hamad Ghaleb. 2022. "Therapeutic Potential of Small Molecules Targeting Oxidative Stress in the Treatment of Chronic Obstructive Pulmonary Disease (COPD): A Comprehensive Review" Molecules 27, no. 17: 5542. https://doi.org/10.3390/molecules27175542

Article Metrics

Back to TopTop